1
|
Gariglio G, Bendova K, Hermann M, Olafsdottir A, Sosabowski JK, Petrik M, von Guggenberg E, Decristoforo C. Comparison of Two Chelator Scaffolds as Basis for Cholecystokinin-2 Receptor Targeting Bimodal Imaging Probes. Pharmaceuticals (Basel) 2024; 17:1569. [PMID: 39770411 PMCID: PMC11676163 DOI: 10.3390/ph17121569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Dual-modality probes, combining positron emission tomography (PET) with fluorescence imaging (FI) capabilities in a single molecule, are of high relevance for the accurate staging and guided resection of tumours. We herein present a pair of candidates targeting the cholecystokinin-2 receptor (CCK2R), namely [68Ga]Ga-CyTMG and [68Ga]Ga-CyFMG. In these probes, the SulfoCy5.5 fluorophore and two units of a CCK2R-binding motif are coupled to the chelator acting as a core scaffold, triazacyclononane-phosphinic acid (TRAP), and Fusarinine C (FSC), respectively. Using this approach, we investigated the influence of these chelators on the final properties. Methods: The synthetic strategy to both precursors was based on the stoichiometric conjugation of the components via click chemistry. The characterization in vitro included the evaluation of the CCK2R affinity and internalization in A431-CCK2R cells. Ex vivo biodistribution as well as PET and FI studies were performed in xenografted mice. Results: 68Ga labelling was accomplished with high radiochemical yield and purity for both precursors. A CCK2R affinity in the subnanomolar range of the conjugates and a receptor-specific uptake of the radioligands in cells were observed. In A431-CCK2R/A431-mock xenografted mice, the investigated compounds showed specific accumulation in the tumours and reduced off-target uptake compared to a previously developed compound. Higher accumulation and prolonged retention in the kidneys were observed for [68Ga]Ga-CyTMG when compared to [68Ga]Ga-CyFMG. Conclusions: Despite the promising targeting properties observed, further probe optimization is required to achieve enhanced imaging contrast at early timepoints. Additionally, the results indicate a distinct influence of the chelators in terms of renal accumulation and retention.
Collapse
Affiliation(s)
- Giacomo Gariglio
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Katerina Bendova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Asta Olafsdottir
- Perceptive Discovery, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Jane K. Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic
- Czech Advanced Technology and Research Institute, Palacky University, 77900 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital, 77900 Olomouc, Czech Republic
| | | | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
2
|
Sun R, Wang Y, Shi W, Zhang H, Liu J, He W. Acidity-Triggered "Sticky Spotlight": CCK2R-Targeted TME-Sensitive NIR Fluorescent Probes for Tumor Imaging In Vivo. Bioconjug Chem 2024; 35:528-539. [PMID: 38514970 DOI: 10.1021/acs.bioconjchem.4c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Cancer which causes high mortality globally threatens public health seriously. There is an urgent need to develop tumor-specific near-infrared (NIR) imaging agents to achieve precise diagnosis and guide effective treatment. In recent years, imaging probes that respond to acidic environments such as endosomes, lysosomes, or acidic tumor microenvironments (TMEs) are being developed. However, because of their nonspecific internalization by both normal and tumor cells, resulting in a poor signal-to-noise ratio in diagnosis, these pH-sensitive probes fail to be applied to in vivo tumor imaging. To address this issue, a cholecystokinin-2 receptor (CCK2R)-targeted TME-sensitive NIR fluorescent probe R2SM was synthesized by coupling pH-sensitive heptamethine cyanine with a CCK2R ligand, minigastrin analogue 11 (MG11) for in vivo imaging, in which MG11 would target overexpressed CCK2Rs in gastrointestinal stromal tumors (GISTs). Cell uptake assay demonstrated that R2SM exhibited a high affinity for CCK2R, leading to receptor-mediated internalization and making probes finally accumulated in the lysosomes of tumor cells, which suggested in the tumor tissues, the probes were distributed in the extracellular acidic TME and intracellular lysosomes. With a pKa of 6.83, R2SM can be activated at the acidic TME (pH = 6.5-6.8) and lysosomes (pH = 4.5-5.0), exhibiting an apparent pH-dependent behavior and generating more intense fluorescence in these acidic environments. In vivo imaging showed that coupling of MG11 with a pH-sensitive NIR probe facilitated the accumulation of probe and enhanced the fluorescence in CCK2R-overexpressed HT-29 tumor cells. A high signal was observed in the tumor region within 0.5 h postinjection, indicating its potential application in intraoperative imaging. Fluorescence imaging of R2SM exhibited higher tumor-to-liver and tumor-to-kidney ratios (2.1:1 and 2.3:1, respectively), compared separately with the probes that are lipophilic, pH-insensitive, or MG11-free. In vitro and in vivo studies demonstrated that the synergistic effect of tumor targeting with pH sensitivity plays a vital role in the high signal-to-noise ratio of the NIR imaging probe. Moreover, different kinds of tumor-targeting vectors could be conjugated simultaneously with the NIR dye, which would further improve the receptor affinity and targeting efficiency.
Collapse
Affiliation(s)
- Ruiqi Sun
- Medical Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuxin Wang
- Medical Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenhui Shi
- Medical Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongfu Zhang
- Medical Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianhua Liu
- Medical Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weina He
- Medical Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
3
|
Happacher I, Aguiar M, Yap A, Decristoforo C, Haas H. Fungal siderophore metabolism with a focus on Aspergillus fumigatus: impact on biotic interactions and potential translational applications. Essays Biochem 2023; 67:829-842. [PMID: 37313590 PMCID: PMC10500206 DOI: 10.1042/ebc20220252] [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: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
Iron is an essential trace element that is limiting in most habitats including hosts for fungal pathogens. Siderophores are iron-chelators synthesized by most fungal species for high-affinity uptake and intracellular handling of iron. Moreover, virtually all fungal species including those lacking siderophore biosynthesis appear to be able to utilize siderophores produced by other species. Siderophore biosynthesis has been shown to be crucial for virulence of several fungal pathogens infecting animals and plants revealing induction of this iron acquisition system during virulence, which offers translational potential of this fungal-specific system. The present article summarizes the current knowledge on the fungal siderophore system with a focus on Aspergillus fumigatus and its potential translational application including noninvasive diagnosis of fungal infections via urine samples, imaging of fungal infections via labeling of siderophores with radionuclides such as Gallium-68 for detection with positron emission tomography, conjugation of siderophores with fluorescent probes, and development of novel antifungal strategies.
Collapse
Affiliation(s)
- Isidor Happacher
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Mario Aguiar
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Annie Yap
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Hubertus Haas
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
4
|
Morath V, Brandt C, Deuschle FC, Mendler CT, Blechert B, Summer D, Barinka C, Decristoforo C, Weber WA, Schwaiger M, Skerra A. Molecular Design of 68Ga- and 89Zr-Labeled Anticalin Radioligands for PET-Imaging of PSMA-Positive Tumors. Mol Pharm 2023; 20:2490-2501. [PMID: 37068305 DOI: 10.1021/acs.molpharmaceut.2c01066] [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: 04/19/2023]
Abstract
Anticalin proteins directed against the prostate-specific membrane antigen (PSMA), optionally having tailored plasma half-life using PASylation technology, show promise as radioligands for PET-imaging of xenograft tumors in mice. To investigate their suitability, the short-circulating unmodified Anticalin was labeled with 68Ga (τ1/2 = 68 min), using the NODAGA chelator, whereas the half-life extended PASylated Anticalin was labeled with 89Zr (τ1/2 = 78 h), using either the linear chelator deferoxamine (Dfo) or a cyclic derivative, fusarinine C (FsC). Different PSMA targeting Anticalin versions (optionally carrying the PASylation sequence) were produced carrying a single exposed N- or C-terminal Cys residue and site-specifically conjugated with the different radiochelators via maleimide chemistry. These protein conjugates were labeled with radioisotopes having distinct physical half-lives and, subsequently, applied for PET-imaging of subcutaneous LNCaP xenograft tumors in CB17 SCID mice. Uptake of the protein tracers into tumor versus healthy tissues was assessed by segmentation of PET data as well as biodistribution analyses. PET-imaging with both the 68Ga-labeled plain Anticalin and the 89Zr-labeled PASylated Anticalin allowed clear delineation of the xenograft tumor. The radioligand A3A5.1-PAS(200)-FsC·89Zr, having an extended plasma half-life, led to a higher tumor uptake 24 h p.i. compared to the 68Ga·NODAGA-Anticalin imaged 60 min p.i. (2.5% ID/g vs 1.2% ID/g). Pronounced demetallation was observed for the 89Zr·Dfo-labeled PASylated Anticalin, which was ∼50% lower in the case of the cyclic radiochelator FsC (p < 0.0001). Adjusting the plasma half-life of Anticalin radioligands using PASylation technology is a viable approach to increase radioisotope accumulation within the tumor. Furthermore, 89Zr-ImmunoPET-imaging using the FsC radiochelator is superior to that using Dfo. Our strategy for the half-life adjustment of a tumor-targeting Anticalin to match the physical half-life of the applied radioisotope illustrates the potential of small binding proteins as an alternative to antibodies for PET-imaging.
Collapse
Affiliation(s)
- Volker Morath
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich 81675, Germany
- Lehrstuhl für Biologische Chemie, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Corinna Brandt
- Lehrstuhl für Biologische Chemie, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Friedrich-Christian Deuschle
- Lehrstuhl für Biologische Chemie, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Claudia T Mendler
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Birgit Blechert
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck 6020, Austria
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology, Czech Academy of Sciences, Vestec 252 50, Czech Republic
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck 6020, Austria
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| |
Collapse
|
5
|
Judmann B, Braun D, Schirrmacher R, Wängler B, Fricker G, Wängler C. Toward the Development of GE11-Based Radioligands for Imaging of Epidermal Growth Factor Receptor-Positive Tumors. ACS OMEGA 2022; 7:27690-27702. [PMID: 35967067 PMCID: PMC9366781 DOI: 10.1021/acsomega.2c03407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
The epidermal growth factor receptor (EGFR) is closely associated with tumor development and progression and thus an important target structure for imaging and therapy of various tumors. As a result of its important role in malignancies of various origins and the fact that antibody-based compounds targeting the EGFR have significant drawbacks in terms of in vivo pharmacokinetics, several attempts have been made within the last five years to develop peptide-based EGFR-specific radioligands based on the GE11 scaffold. However, none of these approaches have shown convincing results so far, which has been proposed to be attributed to different potential challenges associated with the GE11 lead structure: first, an aggregation of radiolabeled peptides, which might prevent their interaction with their target receptor, or second, a relatively low affinity of monomeric GE11, necessitating its conversion into a multimeric or polymeric form to achieve adequate EGFR-targeting properties. In the present work, we investigated if these aforementioned points are indeed critical and if the EGFR-targeting ability of GE11 can be improved by choosing an appropriate hydrophilic molecular design or a peptide multimer system to obtain a promising radiopeptide for the visualization of EGFR-overexpressing malignancies by positron emission tomography (PET). For this purpose, we developed several monovalent 68Ga-labeled GE11-based agents, a peptide homodimer and a homotetramer to overcome the challenges associated with GE11. The developed ligands were successfully labeled with 68Ga3+ in high radiochemical yields of ≥97% and molar activities of 41-104 GBq/μmol. The resulting radiotracers presented log D(7.4) values between -2.17 ± 0.21 and -3.79 ± 0.04 as well as a good stability in human serum with serum half-lives of 112 to 217 min for the monovalent radiopeptides and 84 and 62 min for the GE11 homodimer and homotetramer, respectively. In the following in vitro studies, none of the 68Ga-labeled radiopeptides demonstrated a considerable EGF receptor-specific uptake in EGFR-positive A431 cells. Moreover, none of the agents was able to displace [125I]I-EGF from the EGFR in competitive displacement assays in the same cell line in concentrations of up to 1 mM, whereas the endogenous receptor ligand hEGF demonstrated a high affinity of 15.2 ± 3.3 nM. These results indicate that it is not the aggregation of the GE11 sequence that seems to be the factor limiting the usefulness of the peptide as basis for radiotracer design but the limited affinity of monovalent and small homomultivalent GE11-based radiotracers to the EGFR. This highlights that the development of small-molecule GE11-based radioligands is not promising.
Collapse
Affiliation(s)
- Benedikt Judmann
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Diana Braun
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Department
of Oncology, Division of Oncological Imaging, University of Alberta, 11560 University Avenue, T6G 1Z2 Edmonton, AB, Canada
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Gert Fricker
- Institute
of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
6
|
Usama SM, Marker SC, Hernandez Vargas S, AghaAmiri S, Ghosh SC, Ikoma N, Tran Cao HS, Schnermann MJ, Azhdarinia A. Targeted Dual-Modal PET/SPECT-NIR Imaging: From Building Blocks and Construction Strategies to Applications. Cancers (Basel) 2022; 14:1619. [PMID: 35406390 PMCID: PMC8996983 DOI: 10.3390/cancers14071619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Molecular imaging is an emerging non-invasive method to qualitatively and quantitively visualize and characterize biological processes. Among the imaging modalities, PET/SPECT and near-infrared (NIR) imaging provide synergistic properties that result in deep tissue penetration and up to cell-level resolution. Dual-modal PET/SPECT-NIR agents are commonly combined with a targeting ligand (e.g., antibody or small molecule) to engage biomolecules overexpressed in cancer, thereby enabling selective multimodal visualization of primary and metastatic tumors. The use of such agents for (i) preoperative patient selection and surgical planning and (ii) intraoperative FGS could improve surgical workflow and patient outcomes. However, the development of targeted dual-modal agents is a chemical challenge and a topic of ongoing research. In this review, we define key design considerations of targeted dual-modal imaging from a topological perspective, list targeted dual-modal probes disclosed in the last decade, review recent progress in the field of NIR fluorescent probe development, and highlight future directions in this rapidly developing field.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Sierra C. Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Sukhen C. Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (N.I.); (H.S.T.C.)
| | - Hop S. Tran Cao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (N.I.); (H.S.T.C.)
| | - Martin J. Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| |
Collapse
|
7
|
Zheng F, Huang X, Ding J, Bi A, Wang S, Chen F, Zeng W. NIR-I Dye-Based Probe: A New Window for Bimodal Tumor Theranostics. Front Chem 2022; 10:859948. [PMID: 35402374 PMCID: PMC8984032 DOI: 10.3389/fchem.2022.859948] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Near-infrared (NIR, 650-1700 nm) bioimaging has emerged as a powerful strategy in tumor diagnosis. In particular, NIR-I fluorescence imaging (650-950 nm) has drawn more attention, benefiting from the high quantum yield and good biocompatibility. Since their biomedical applications are slightly limited by their relatively low penetration depth, NIR-I fluorescence imaging probes have been under extensive development in recent years. This review summarizes the particular application of the NIR-I fluorescent dye-contained bimodal probes, with emphasis on related nanoprobes. These probes have enabled us to overcome the drawbacks of individual imaging modalities as well as achieve synergistic imaging. Meanwhile, the application of these NIR-I fluorescence-based bimodal probes for cancer theranostics is highlighted.
Collapse
Affiliation(s)
- Fan Zheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Xueyan Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Jipeng Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Anyao Bi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Shifen Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Fei Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| |
Collapse
|
8
|
[ 68Ga]Ga-DFO-c(RGDyK): Synthesis and Evaluation of Its Potential for Tumor Imaging in Mice. Int J Mol Sci 2021; 22:ijms22147391. [PMID: 34299008 PMCID: PMC8306578 DOI: 10.3390/ijms22147391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 11/30/2022] Open
Abstract
Angiogenesis has a pivotal role in tumor growth and the metastatic process. Molecular imaging was shown to be useful for imaging of tumor-induced angiogenesis. A great variety of radiolabeled peptides have been developed to target αvβ3 integrin, a target structure involved in the tumor-induced angiogenic process. The presented study aimed to synthesize deferoxamine (DFO)-based c(RGD) peptide conjugate for radiolabeling with gallium-68 and perform its basic preclinical characterization including testing of its tumor-imaging potential. DFO-c(RGDyK) was labeled with gallium-68 with high radiochemical purity. In vitro characterization including stability, partition coefficient, protein binding determination, tumor cell uptake assays, and ex vivo biodistribution as well as PET/CT imaging was performed. [68Ga]Ga-DFO-c(RGDyK) showed hydrophilic properties, high stability in PBS and human serum, and specific uptake in U-87 MG and M21 tumor cell lines in vitro and in vivo. We have shown here that [68Ga]Ga-DFO-c(RGDyK) can be used for αvβ3 integrin targeting, allowing imaging of tumor-induced angiogenesis by positron emission tomography.
Collapse
|
9
|
Simões JCS, Sarpaki S, Papadimitroulas P, Therrien B, Loudos G. Conjugated Photosensitizers for Imaging and PDT in Cancer Research. J Med Chem 2020; 63:14119-14150. [PMID: 32990442 DOI: 10.1021/acs.jmedchem.0c00047] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early cancer detection and perfect understanding of the disease are imperative toward efficient treatments. It is straightforward that, for choosing a specific cancer treatment methodology, diagnostic agents undertake a critical role. Imaging is an extremely intriguing tool since it assumes a follow up to treatments to survey the accomplishment of the treatment and to recognize any conceivable repeating injuries. It also permits analysis of the disease, as well as to pursue treatment and monitor the possible changes that happen on the tumor. Likewise, it allows screening the adequacy of treatment and visualizing the state of the tumor. Additionally, when the treatment is finished, observing the patient is imperative to evaluate the treatment methodology and adjust the treatment if necessary. The goal of this review is to present an overview of conjugated photosensitizers for imaging and therapy.
Collapse
Affiliation(s)
- João C S Simões
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland.,BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | - Sophia Sarpaki
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | | | - Bruno Therrien
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland
| | - George Loudos
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| |
Collapse
|
10
|
Pfister J, Lichius A, Summer D, Haas H, Kanagasundaram T, Kopka K, Decristoforo C. Live-cell imaging with Aspergillus fumigatus-specific fluorescent siderophore conjugates. Sci Rep 2020; 10:15519. [PMID: 32968138 PMCID: PMC7511942 DOI: 10.1038/s41598-020-72452-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Live-cell imaging allows the in vivo analysis of subcellular localisation dynamics of physiological processes with high spatial-temporal resolution. However, only few fluorescent dyes have been custom-designed to facilitate species-specific live-cell imaging approaches in filamentous fungi to date. Therefore, we developed fluorescent dye conjugates based on the sophisticated iron acquisition system of Aspergillus fumigatus by chemical modification of the siderophore triacetylfusarinine C (TAFC). Various fluorophores (FITC, NBD, Ocean Blue, BODIPY 630/650, SiR, TAMRA and Cy5) were conjugated to diacetylfusarinine C (DAFC). Gallium-68 labelling enabled in vitro and in vivo characterisations. LogD, uptake assays and growth assays were performed and complemented by live-cell imaging in different Aspergillus species. Siderophore conjugates were specifically recognised by the TAFC transporter MirB and utilized as an iron source in growth assays. Fluorescence microscopy revealed uptake dynamics and differential subcellular accumulation patterns of all compounds inside fungal hyphae.[Fe]DAFC-NBD and -Ocean Blue accumulated in vacuoles, whereas [Fe]DAFC-BODIPY, -SiR and -Cy5 localised to mitochondria. [Fe]DAFC -FITC showed a uniform cytoplasmic distribution, whereas [Fe]DAFC-TAMRA was not internalised at all. Co-staining experiments with commercially available fluorescent dyes confirmed these findings. Overall, we developed a new class of fluorescent dyes that vary in intracellular fungal targeting , thereby providing novel tools for live-cell imaging applications for Aspergillus fumigatus.
Collapse
Affiliation(s)
- Joachim Pfister
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Alexander Lichius
- Department of Microbiology, University Innsbruck, Innsbruck, Austria
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Hubertus Haas
- Division of Molecular Biology, Medical University Innsbruck, Innsbruck, Austria
| | - Thines Kanagasundaram
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center, (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center, (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
11
|
Summer D, Petrik M, Mayr S, Hermann M, Kaeopookum P, Pfister J, Klingler M, Rangger C, Haas H, Decristoforo C. Hybrid Imaging Agents for Pretargeting Applications Based on Fusarinine C-Proof of Concept. Molecules 2020; 25:E2123. [PMID: 32370017 PMCID: PMC7249120 DOI: 10.3390/molecules25092123] [Citation(s) in RCA: 4] [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: 04/17/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022] Open
Abstract
Hybrid imaging combining the beneficial properties of radioactivity and optical imaging within one imaging probe has gained increasing interest in radiopharmaceutical research. In this study, we modified the macrocyclic gallium-68 chelator fusarinine C (FSC) by conjugating a fluorescent moiety and tetrazine (Tz) moieties. The resulting hybrid imaging agents were used for pretargeting applications utilizing click reactions with a trans-cyclooctene (TCO) tagged targeting vector for a proof of principle both in vitro and in vivo. Starting from FSC, the fluorophores Sulfocyanine-5, Sulfocyanine-7, or IRDye800CW were conjugated, followed by introduction of one or two Tz motifs, resulting in mono and dimeric Tz conjugates. Evaluation included fluorescence microscopy, binding studies, logD, protein binding, in vivo biodistribution, µPET (micro-positron emission tomography), and optical imaging (OI) studies. 68Ga-labeled conjugates showed suitable hydrophilicity, high stability, and specific targeting properties towards Rituximab-TCO pre-treated CD20 expressing Raji cells. Biodistribution studies showed fast clearance and low accumulation in non-targeted organs for both SulfoCy5- and IRDye800CW-conjugates. In an alendronate-TCO based bone targeting model the dimeric IRDye800CW-conjugate resulted in specific targeting using PET and OI, superior to the monomer. This proof of concept study showed that the preparation of FSC-Tz hybrid imaging agents for pretargeting applications is feasible, making such compounds suitable for hybrid imaging applications.
Collapse
Affiliation(s)
- Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 772-00 Olomouc, Czech Republic;
| | - Sonja Mayr
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| | - Martin Hermann
- Department of Anaesthesia and Intensive Care, Medical University Innsbruck, A-6020 Innsbruck, Austria;
| | - Piriya Kaeopookum
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| | - Joachim Pfister
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| | - Maximilian Klingler
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| | - Hubertus Haas
- Institute of Molecular Biology, Medical University Innsbruck, A-6020 Innsbruck, Austria;
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (D.S.); (S.M.); (P.K.); (J.P.); (M.K.); (C.R.)
| |
Collapse
|
12
|
Pfister J, Summer D, Petrik M, Khoylou M, Lichius A, Kaeopookum P, Kochinke L, Orasch T, Haas H, Decristoforo C. Hybrid Imaging of Aspergillus fumigatus Pulmonary Infection with Fluorescent, 68Ga-Labelled Siderophores. Biomolecules 2020; 10:E168. [PMID: 31979017 PMCID: PMC7072563 DOI: 10.3390/biom10020168] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/19/2020] [Indexed: 01/16/2023] Open
Abstract
Aspergillus fumigatus (A. fumigatus) is a human pathogen causing severe invasive fungal infections, lacking sensitive and selective diagnostic tools. A. fumigatus secretes the siderophore desferri-triacetylfusarinine C (TAFC) to acquire iron from the human host. TAFC can be labelled with gallium-68 to perform positron emission tomography (PET/CT) scans. Here, we aimed to chemically modify TAFC with fluorescent dyes to combine PET/CT with optical imaging for hybrid imaging applications. Starting from ferric diacetylfusarinine C ([Fe]DAFC), different fluorescent dyes were conjugated (Cy5, SulfoCy5, SulfoCy7, IRDye 800CW, ATTO700) and labelled with gallium-68 for in vitro and in vivo characterisation. Uptake assays, growth assays and live-cell imaging as well as biodistribution, PET/CT and ex vivo optical imaging in an infection model was performed. Novel fluorophore conjugates were recognized by the fungal TAFC transporter MirB and could be utilized as iron source. Fluorescence microscopy showed partial accumulation into hyphae. µPET/CT scans of an invasive pulmonary aspergillosis (IPA) rat model revealed diverse biodistribution patterns for each fluorophore. [68Ga]Ga-DAFC-Cy5/SufloCy7 and -IRDye 800CW lead to a visualization of the infected region of the lung. Optical imaging of ex vivo lungs corresponded to PET images with high contrast of infection versus non-infected areas. Although fluorophores had a decisive influence on targeting and pharmacokinetics, these siderophores have potential as a hybrid imaging compounds combining PET/CT with optical imaging applications.
Collapse
Affiliation(s)
- Joachim Pfister
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (J.P.); (D.S.); (P.K.); (L.K.)
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (J.P.); (D.S.); (P.K.); (L.K.)
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 772-00 Olomouc, Czech Republic; (M.P.); (M.K.)
| | - Marta Khoylou
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 772-00 Olomouc, Czech Republic; (M.P.); (M.K.)
| | - Alexander Lichius
- Department of Microbiology, University Innsbruck, A-6020 Innsbruck, Austria;
| | - Piriya Kaeopookum
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (J.P.); (D.S.); (P.K.); (L.K.)
| | - Laurin Kochinke
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (J.P.); (D.S.); (P.K.); (L.K.)
| | - Thomas Orasch
- Division of Molecular Biology, Medical University Innsbruck, A-6020 Innsbruck, Austria; (T.O.); (H.H.)
| | - Hubertus Haas
- Division of Molecular Biology, Medical University Innsbruck, A-6020 Innsbruck, Austria; (T.O.); (H.H.)
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria; (J.P.); (D.S.); (P.K.); (L.K.)
| |
Collapse
|
13
|
Kaeopookum P, Summer D, Pfister J, Orasch T, Lechner BE, Petrik M, Novy Z, Matuszczak B, Rangger C, Haas H, Decristoforo C. Modifying the Siderophore Triacetylfusarinine C for Molecular Imaging of Fungal Infection. Mol Imaging Biol 2019; 21:1097-1106. [PMID: 30838551 PMCID: PMC6877352 DOI: 10.1007/s11307-019-01325-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Aspergillus fumigatus produces the siderophore triacetylfusarinine C (TAFC) for iron acquisition which is essential for its virulence. Therefore, TAFC is a specific marker for invasive aspergillosis. We have shown previously that positron emission tomography (PET) imaging with [68Ga]TAFC exhibited excellent targeting properties in an A. fumigatus rat infection model. In this study, we aimed to prepare TAFC analogs modifying fusarinine C (FSC) by acylation with different carbon chain lengths as well as with charged substituents and investigated the influence of introduced substituents on preservation of TAFC characteristics in vitro and in vivo. PROCEDURES Fifteen TAFC derivatives were prepared and labeled with gallium-68. In vitro uptake assays were carried out in A. fumigatus under iron-replete as well as iron-depleted conditions and distribution coefficient was determined. Based on these assays, three compounds, [68Ga]tripropanoyl(FSC) ([68Ga]TPFC), [68Ga]diacetylbutanoyl(FSC) ([68Ga]DABuFC), and [68Ga]trisuccinyl(FSC) ([68Ga]FSC(suc)3), with high, medium, and low in vitro uptake in fungal cultures, were selected for further evaluation. Stability and protein binding were evaluated and in vivo imaging performed in the A. fumigatus rat infection model. RESULTS In vitro uptake studies using A. fumigatus revealed specific uptake of mono- and trisubstituted TAFC derivatives at RT. Lipophilicities as expressed by logD were 0.34 to - 3.80. The selected compounds displayed low protein binding and were stable in PBS and serum. Biodistribution and image contrast in PET/X-ray computed tomography of [68Ga]TPFC and [68Ga]DABuFC were comparable to [68Ga]TAFC, whereas no uptake in the infected region was observed with [68Ga]FSC(suc)3. CONCLUSIONS Our studies show the possibility to modify TAFC without losing its properties and specific recognition by A. fumigatus. This opens also new ways for multimodality imaging or theranostics of fungal infection by introducing functionalities such as fluorescent dyes or antifungal moieties.
Collapse
Affiliation(s)
- Piriya Kaeopookum
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
- Research and Development Division, Thailand Institute of Nuclear Technology, Nakhon Nayok, Thailand
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Joachim Pfister
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Orasch
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Beatrix E Lechner
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Zbynek Novy
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Barbara Matuszczak
- Institute of Pharmacy, Pharmaceutical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Hubertus Haas
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
14
|
Kaeopookum P, Petrik M, Summer D, Klinger M, Zhai C, Rangger C, Haubner R, Haas H, Hajduch M, Decristoforo C. Comparison of 68Ga-labeled RGD mono- and multimers based on a clickable siderophore-based scaffold. Nucl Med Biol 2019; 78-79:1-10. [PMID: 31678781 DOI: 10.1016/j.nucmedbio.2019.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Cyclic pentapeptides containing the amino acid sequence arginine-glycine-aspartic (RGD) have been widely applied to target αvβ3 integrin, which is upregulated in various tumors during tumor-induced angiogenesis. Multimeric cyclic RGD peptides have been reported to be advantageous over monomeric counterparts for angiogenesis imaging. Here, we prepared mono-, di-, and trimeric cyclic arginine-glycine-aspartic-D-phenylalanine-lysine (c (RGDfK)) derivatives by conjugation with the natural chelator fusarinine C (FSC) using click chemistry based on copper (I)-catalyzed azide-alkyne cycloaddition (CuAAC). The αvβ3 binding properties of 68Ga-labeled mono-, di-, and trimeric c(RGDfK) peptides were evaluated in vitro as well as in vivo and compared with the references monomeric [68Ga]GaNODAGA-c(RGDfK) and trimeric [68Ga]GaFSC(suc-c(RGDfK))3. All 68Ga-labeled c(RGDfK) peptides displayed hydrophilicity (logD = -2.96 to -3.80), low protein binding and were stable in phosphate buffered-saline (PBS) and serum up to 2 h. In vitro internalization assays with human melanoma M21 (αvβ3-positive) and M21-L (αvβ3-negative) cell lines showed specific uptake of all derivatives and increased in the series: mono- < di- < trimeric peptide. The highest tumor uptake, tumor-to-background ratios, and image contrast were found for the dimeric [68Ga]GaMAFC(c(RGDfK)aza)2. In conclusion, we developed a novel strategy for direct, straight forward preparation of mono-, di-, and trimeric c(RGDfK) conjugates based on the FSC scaffold. Interestingly, the best αvβ3 imaging properties were found for the dimeric [68Ga]GaMAFC(c(RGDfK)aza)2.
Collapse
Affiliation(s)
- Piriya Kaeopookum
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria; Research and Development Division, Thailand Institute of Nuclear Technology, Nakhon Nayok, Thailand
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Maximilian Klinger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Chuangyan Zhai
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria; School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Hubertus Haas
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
15
|
Wan W, Silva MS, McMillen CD, Creager SE, Smith RC. Highly Luminescent Heavier Main Group Analogues of Boron-Dipyrromethene. J Am Chem Soc 2019; 141:8703-8707. [PMID: 31120244 DOI: 10.1021/jacs.9b03235] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The preparation and photophysical properties of two heavier main group element analogues of boron-dipyrromethene (BODIPY) chromophores are described. Specifically, we have prepared dipyrrin complexes of dichlorogallate (GADIPY) or phenylphosphenium (PHODIPY) units. Whereas cationic PHODIPY is labile, decomposing to a phosphine over time, GADIPY is readily prepared in good yield as a crystalline solid having moderate air- and water-stability. Crystallographically characterized GADIPY displays intense green photoluminescence (λem = 505 nm, Φem = 0.91 in toluene). These inaugural heavier main group element analogues of BODIPY offer a glimpse into the potential for elaboration to a panoply of chromophores with diverse photophysical properties.
Collapse
Affiliation(s)
- Wang Wan
- Department of Chemistry , Clemson University , Clemson , South Carolina 29634 , United States
| | - Mayura S Silva
- Department of Chemistry , Clemson University , Clemson , South Carolina 29634 , United States
| | - Colin D McMillen
- Department of Chemistry , Clemson University , Clemson , South Carolina 29634 , United States
| | - Stephen E Creager
- Department of Chemistry , Clemson University , Clemson , South Carolina 29634 , United States.,Center for Optical Materials Science and Engineering Technology, Clemson University , Anderson , South Carolina 29625 , United States
| | - Rhett C Smith
- Department of Chemistry , Clemson University , Clemson , South Carolina 29634 , United States.,Center for Optical Materials Science and Engineering Technology, Clemson University , Anderson , South Carolina 29625 , United States
| |
Collapse
|
16
|
Hernandez Vargas S, Ghosh SC, Azhdarinia A. New Developments in Dual-Labeled Molecular Imaging Agents. J Nucl Med 2019; 60:459-465. [PMID: 30733318 DOI: 10.2967/jnumed.118.213488] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
Intraoperative detection of tumors has had a profound impact on how cancer surgery is performed and addresses critical unmet needs in surgical oncology. Tumor deposits, margins, and residual cancer can be imaged through the use of fluorescent contrast agents during surgical procedures to complement visual and tactile guidance. The combination of fluorescent and nuclear contrast into a multimodality agent builds on these capabilities by adding quantitative, noninvasive nuclear imaging capabilities to intraoperative imaging. This review focuses on new strategies for the development and evaluation of targeted dual-labeled molecular imaging agents while highlighting the successful first-in-human application of this technique.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sukhen C Ghosh
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Ali Azhdarinia
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
17
|
Pretargeted Imaging with Gallium-68-Improving the Binding Capability by Increasing the Number of Tetrazine Motifs. Pharmaceuticals (Basel) 2018; 11:ph11040102. [PMID: 30314332 PMCID: PMC6316846 DOI: 10.3390/ph11040102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/29/2023] Open
Abstract
The inverse electron-demand Diels-Alder reaction between 1,2,4,5-tetrazine (Tz) and trans-cyclooct-2-ene (TCO) has gained increasing attraction among extensive studies on click chemistry due to its exceptionally fast reaction kinetics and high selectivity for in vivo pretargeting applications including PET imaging. The facile two-step approach utilizing TCO-modified antibodies as targeting structures has not made it into clinics yet. An increase in the blood volume of humans in comparison to mice seems to be the major limitation. This study aims to show if the design of multimeric Tz-ligands by chelator scaffolding can improve the binding capacity and may lead to enhanced PET imaging with gallium-68. We utilized for this purpose the macrocyclic siderophore Fusarinine C (FSC) which allows conjugation of up to three Tz-residues due to three primary amines available for site specific modification. The resulting mono- di- and trimeric conjugates were radiolabelled with gallium-68 and characterized in vitro (logD, protein binding, stability, binding towards TCO modified rituximab (RTX)) and in vivo (biodistribution- and imaging studies in normal BALB/c mice using a simplified RTX-TCO tumour surrogate). The 68Ga-labelled FSC-based Tz-ligands showed suitable hydrophilicity, high stability and high targeting specificity. The binding capacity to RTX-TCO was increased according to the grade of multimerization. Corresponding in vivo studies showed a multimerization typical profile but generally suitable pharmacokinetics with low accumulation in non-targeted tissue. Imaging studies in RTX-TCO tumour surrogate bearing BALB/c mice confirmed this trend and revealed improved targeting by multimerization as increased accumulation in RTX-TCO positive tissue was observed.
Collapse
|
18
|
Multimerization results in formation of re-bindable metabolites: A proof of concept study with FSC-based minigastrin imaging probes targeting CCK2R expression. PLoS One 2018; 13:e0201224. [PMID: 30059514 PMCID: PMC6066219 DOI: 10.1371/journal.pone.0201224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/11/2018] [Indexed: 12/21/2022] Open
Abstract
Positron emission tomography (PET) with radiolabelled peptide-based tracers has attracted great interest in oncology over the past decades. The success of imaging is closely related to sufficient uptake of the radiotracer in malignant tissue and for this sufficient biological half-life, particularly in the bloodstream, is mandatory. Fast enzymatic degradation during circulation leading to insufficient imaging abilities of peptide-based radioligands remains a major issue. The design of multimeric constructs, bearing multiple targeting moieties, has been widely applied to improve target interaction. This concept may also be applied to prolong the biological half-life of peptide-based radiopharmaceuticals as enzymatic degradation can result in formation of metabolites still capable to interact with the target binding site. In this study we aimed to identify such metabolites and therefore we utilized the siderophore-based bifunctional chelator fusarinine C (FSC) for the design of novel mono- and multimeric constructs, bearing minigastrin (MG) analogues as targeting moieties to address cholecystokinin-2 receptors (CCK2R) which are overexpressed in a variety of cancerous diseases and are well known for fast enzymatic degradation, particularly for truncated des-(Glu)5-MG members, such as MG11. FSC-based imaging probes were radiolabelled with gallium-68 and characterized in vitro (logD, protein binding, affinity and cell-uptake studies, stability and metabolite studies, as well as generation of corresponding metabolites by artificial enzymatic degradation) and in vivo (biodistribution in A431-CCK2R/A431-mock tumour xenografted BALB/c nude mice and stability in blood of living BALB/c mice and analysis of corresponding organ homogenates and urine to identify degradation products). In summary, multimerization was accompanied by partial improvement towards targeting abilities. Identified metabolites formed by artificial enzymatic cleavage of trimeric FSC-MG conjugates in vitro contained intact binding sequences for the receptor. Furthermore, the 68Ga-labelled trimers exhibiting increasing uptake of radioligand in tumour tissue over time and improved in vivo stability in blood samples of living animals of the trimers compared to corresponding mono- and dimers, strongly supporting our hypothesis.
Collapse
|
19
|
Ahn SH, Boros E. Nuclear and Optical Bimodal Imaging Probes Using Sequential Assembly: A Perspective. Cancer Biother Radiopharm 2018; 33:308-315. [PMID: 30004803 DOI: 10.1089/cbr.2018.2499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
New, targeted imaging tracers enable improved diagnosis, staging, and planning of treatment of disease and represent an important step toward personalized medicine applications. The combination of radioisotopes for nuclear imaging with fluorophores for fluorescence imaging provides the possibility to noninvasively assess disease burden in a patient using positron emission tomography/single-photon emission computed tomography, followed by fluorescence imaging-assisted surgical intervention in close succession. Probes enabling imaging with both modalities pose a design, synthesis, and pharmacokinetics challenge. In this study, the authors strive to summarize recent efforts toward optimized, discrete, bimodal probes as well as a perspective on future directions of this burgeoning subfield of targeted imaging probe development.
Collapse
Affiliation(s)
- Shin Hye Ahn
- Department of Chemistry, Stony Brook University , Stony Brook, New York
| | - Eszter Boros
- Department of Chemistry, Stony Brook University , Stony Brook, New York
| |
Collapse
|
20
|
Zhao J, Chen J, Ma S, Liu Q, Huang L, Chen X, Lou K, Wang W. Recent developments in multimodality fluorescence imaging probes. Acta Pharm Sin B 2018; 8:320-338. [PMID: 29881672 PMCID: PMC5989919 DOI: 10.1016/j.apsb.2018.03.010] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Multimodality optical imaging probes have emerged as powerful tools that improve detection sensitivity and accuracy, important in disease diagnosis and treatment. In this review, we focus on recent developments of optical fluorescence imaging (OFI) probe integration with other imaging modalities such as X-ray computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), single-photon emission computed tomography (SPECT), and photoacoustic imaging (PAI). The imaging technologies are briefly described in order to introduce the strengths and limitations of each techniques and the need for further multimodality optical imaging probe development. The emphasis of this account is placed on how design strategies are currently implemented to afford physicochemically and biologically compatible multimodality optical fluorescence imaging probes. We also present studies that overcame intrinsic disadvantages of each imaging technique by multimodality approach with improved detection sensitivity and accuracy.
Collapse
Affiliation(s)
- Jianhong Zhao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Junwei Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Shengnan Ma
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Qianqian Liu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Lixian Huang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Xiani Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyan Lou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131-0001, USA
| |
Collapse
|
21
|
Exploiting the Concept of Multivalency with 68Ga- and 89Zr-Labelled Fusarinine C-Minigastrin Bioconjugates for Targeting CCK2R Expression. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:3171794. [PMID: 29849512 PMCID: PMC5914118 DOI: 10.1155/2018/3171794] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/20/2018] [Indexed: 11/17/2022]
Abstract
Cholecystokinin-2 receptors (CCK2R) are overexpressed in a variety of malignant diseases and therefore have gained certain attention for peptide receptor radionuclide imaging. Among extensive approaches to improve pharmacokinetics and metabolic stability of minigastrin (MG) based radioligands, the concept of multivalency for enhanced tumour targeting has not been investigated extensively. We therefore utilized fusarinine C (FSC) as chelating scaffold for novel mono-, di-, and trimeric bioconjugates for targeting CCK2R expression. FSC-based imaging probes were radiolabelled with positron emitting radionuclides (gallium-68 and zirconium-89) and characterized in vitro (logD, IC50, and cell uptake) and in vivo (metabolic stability in BALB/c mice, biodistribution profile, and microPET/CT imaging in A431-CCK2R/A431-mock tumour xenografted BALB/c nude mice). Improved targeting did not fully correlate with the grade of multimerization. The divalent probe showed higher receptor affinity and increased CCK2R mediated cell uptake while the trimer remained comparable to the monomer. In vivo biodistribution studies 1 h after administration of the 68Ga-labelled radioligands confirmed this trend, but imaging at late time point (24 h) with 89Zr-labelled counterparts showed a clearly enhanced imaging contrast of the trimeric probe compared to the mono- and dimer. Furthermore, in vivo stability studies showed a higher metabolic stability for multimeric probes compared to the monomeric bioconjugate. In summary, we could show that FSC can be utilized as suitable scaffold for novel mono- and multivalent imaging probes for CCK2R-related malignancies with partly improved targeting properties for multivalent conjugates. The increased tumour accumulation of the trimer 24 h postinjection (p.i.) can be explained by slower clearance and increased metabolic stability of multimeric conjugates.
Collapse
|
22
|
Summer D, Garousi J, Oroujeni M, Mitran B, Andersson KG, Vorobyeva A, Löfblom J, Orlova A, Tolmachev V, Decristoforo C. Cyclic versus Noncyclic Chelating Scaffold for 89Zr-Labeled ZEGFR:2377 Affibody Bioconjugates Targeting Epidermal Growth Factor Receptor Overexpression. Mol Pharm 2017; 15:175-185. [PMID: 29160082 PMCID: PMC5751887 DOI: 10.1021/acs.molpharmaceut.7b00787] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
![]()
Zirconium-89
is an emerging radionuclide for positron emission
tomography (PET) especially for biomolecules with slow pharmacokinetics
as due to its longer half-life, in comparison to fluorine-18 and gallium-68,
imaging at late time points is feasible. Desferrioxamine B (DFO),
a linear bifunctional chelator (BFC) is mostly used for this radionuclide
so far but shows limitations regarding stability. Our group recently
reported on fusarinine C (FSC) with similar zirconium-89 complexing
properties but potentially higher stability related to its cyclic
structure. This study was designed to compare FSC and DFO head-to-head
as bifunctional chelators for 89Zr-radiolabeled EGFR-targeting
ZEGFR:2377 affibody bioconjugates. FSC-ZEGFR:2377 and DFO-ZEGFR:2377
were evaluated regarding radiolabeling, in vitro stability,
specificity, cell uptake, receptor affinity, biodistribution, and
microPET-CT imaging. Both conjugates were efficiently labeled with
zirconium-89 at room temperature but radiochemical yields increased
substantially at elevated temperature, 85 °C. Both 89Zr-FSC-ZEGFR:2377 and 89Zr-DFO-ZEGFR:2377 revealed remarkable
specificity, affinity and slow cell-line dependent internalization.
Radiolabeling at 85 °C showed comparable results in A431 tumor
xenografted mice with minor differences regarding blood clearance,
tumor and liver uptake. In comparison 89Zr-DFO-ZEGFR:2377,
radiolabeled at room temperature, showed a significant difference
regarding tumor-to-organ ratios. MicroPET-CT imaging studies of 89Zr-FSC-ZEGFR:2377 as well as 89Zr-DFO-ZEGFR:2377
confirmed these findings. In summary we were able to show that FSC
is a suitable alternative to DFO for radiolabeling of biomolecules
with zirconium-89. Furthermore, our findings indicate that 89Zr-radiolabeling of DFO conjugates at higher temperature reduces
off-chelate binding leading to significantly improved tumor-to-organ
ratios and therefore enhancing image contrast.
Collapse
Affiliation(s)
- Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Javad Garousi
- Institute of Immunology, Genetic and Pathology, Uppsala University , SE-75185 Uppsala, Sweden
| | - Maryam Oroujeni
- Institute of Immunology, Genetic and Pathology, Uppsala University , SE-75185 Uppsala, Sweden
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University , SE-751 83 Uppsala, Sweden
| | - Ken G Andersson
- Division of Protein Technology, KTH Royal Institute of Technology , SE-10691 Stockholm, Sweden
| | - Anzhelika Vorobyeva
- Institute of Immunology, Genetic and Pathology, Uppsala University , SE-75185 Uppsala, Sweden
| | - John Löfblom
- Division of Protein Technology, KTH Royal Institute of Technology , SE-10691 Stockholm, Sweden
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University , SE-751 83 Uppsala, Sweden
| | - Vladimir Tolmachev
- Institute of Immunology, Genetic and Pathology, Uppsala University , SE-75185 Uppsala, Sweden
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| |
Collapse
|