51
|
Palmioli A, Nicolini G, Tripodi F, Orsato A, Ceresa C, Donzelli E, Arici M, Coccetti P, Rocchetti M, La Ferla B, Airoldi C. Targeting GRP receptor: Design, synthesis and preliminary biological characterization of new non-peptide antagonists of bombesin. Bioorg Chem 2021; 109:104739. [PMID: 33626451 DOI: 10.1016/j.bioorg.2021.104739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 01/29/2023]
Abstract
We report the rational design, synthesis, and in vitro preliminary evaluation of a new small library of non-peptide ligands of Gastrin Releasing Peptide Receptor (GRP-R), able to antagonize its natural ligand bombesin (BN) in the nanomolar range of concentration. GRP-R is a transmembrane G-protein coupled receptor promoting the stimulation of cancer cell proliferation. Being overexpressed on the surface of different human cancer cell lines, GRP-R is ideal for the selective delivery to tumor cells of both anticancer drug and diagnostic devices. What makes very challenging the design of non-peptide BN analogues is that the 3D structure of the GRP-R is not available, which is the case for many membrane-bound receptors. Thus, the design of GRP-R ligands has to be based on the structure of its natural ligands, BN and GRP. We recently mapped the BN binding epitope by NMR and here we exploited the same spectroscopy, combined with MD, to define BN conformation in proximity of biological membranes, where the interaction with GRP-R takes place. The gained structural information was used to identify a rigid C-galactosidic scaffold able to support pharmacophore groups mimicking the BN key residues' side chains in a suitable manner for binding to GRP-R. Our BN antagonists represent hit compounds for the rational design and synthesis of new ligands and modulators of GRP-R. The further optimization of the pharmacophore groups will allow to increase the biological activity. Due to their favorable chemical properties and stability, they could be employed for the active receptor-mediated targeting of GRP-R positive tumors.
Collapse
Affiliation(s)
- Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy; Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy
| | - Gabriella Nicolini
- Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy; School of Medicine and Surgery, Experimental Neurology Unit, University of Milano - Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | - Farida Tripodi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Alexandre Orsato
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy; Departamento de Química, CCE, Universidade Estadual de Londrina, CEP 86057-970 Londrina, Paraná, Brazil
| | - Cecilia Ceresa
- Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy; School of Medicine and Surgery, Experimental Neurology Unit, University of Milano - Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | - Elisabetta Donzelli
- Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy; School of Medicine and Surgery, Experimental Neurology Unit, University of Milano - Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | - Martina Arici
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Paola Coccetti
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy.
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy; Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy.
| |
Collapse
|
52
|
Rinne SS, Abouzayed A, Gagnon K, Tolmachev V, Orlova A. 66Ga-PET-imaging of GRPR-expression in prostate cancer: production and characterization of [ 66Ga]Ga-NOTA-PEG 2-RM26. Sci Rep 2021; 11:3631. [PMID: 33574368 PMCID: PMC7878787 DOI: 10.1038/s41598-021-82995-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
Molecular imaging of the gastrin-releasing peptide receptor (GRPR) could improve patient management in prostate cancer. This study aimed to produce gallium-66 (T½ = 9.5 h) suitable for radiolabeling, and investigate the imaging properties of gallium-66 labeled GRPR-antagonist NOTA-PEG2-RM26 for later-time point PET-imaging of GRPR expression. Gallium-66 was cyclotron-produced using a liquid target, and enriched [66Zn]Zn(NO3)2. In vitro, [66Ga]Ga-NOTA-PEG2-RM26 was characterized in GRPR-expressing PC-3 prostate cancer cells. In vivo, specificity test and biodistribution studies were performed 3 h and 22 h pi in PC-3 xenografted mice. microPET/MR was performed 3 h and 22 h pi. Biodistribution of [66Ga]Ga-NOTA-PEG2-RM26 was compared with [68Ga]Ga-NOTA-PEG2-RM26 3 h pi. [66Ga]Ga-NOTA-PEG2-RM26 was successfully prepared with preserved binding specificity and high affinity towards GRPR. [66Ga]Ga-NOTA-PEG2-RM26 cleared rapidly from blood via kidneys. Tumor uptake was GRPR-specific and exceeded normal organ uptake. Normal tissue clearance was limited, resulting in no improvement of tumor-to-organ ratios with time. Tumors could be clearly visualized using microPET/MR. Gallium-66 was successfully produced and [66Ga]Ga-NOTA-PEG2-RM26 was able to clearly visualize GRPR-expression both shortly after injection and on the next day using PET. However, delayed imaging did not improve contrast for Ga-labeled NOTA-PEG2-RM26.
Collapse
Affiliation(s)
- Sara S Rinne
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, Tomsk, Russia.
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
53
|
Mitran B, Tolmachev V, Orlova A. Radiolabeled GRPR Antagonists for Imaging of Disseminated Prostate Cancer - Influence of Labeling Chemistry on Targeting Properties. Curr Med Chem 2021; 27:7090-7111. [PMID: 32164503 DOI: 10.2174/0929867327666200312114902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/31/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Radionuclide molecular imaging of Gastrin-Releasing Peptide Receptor (GRPR) expression promises unparalleled opportunities for visualizing subtle prostate tumors, which due to small size, adjacent benign tissue, or a challenging location would otherwise remain undetected by conventional imaging. Achieving high imaging contrast is essential for this purpose and the molecular design of any probe for molecular imaging of prostate cancer should be aimed at obtaining as high tumor-to-organ ratios as possible. OBJECTIVE This short review summarizes the key imaging modalities currently used in prostate cancer, with a special focus on radionuclide molecular imaging. Emphasis is laid mainly on the issue of radiometals labeling chemistry and its influence on the targeting properties and biodistribution of radiolabeled GRPR antagonists for imaging of disseminated prostate cancer. METHODS A comprehensive literature search of the PubMed/MEDLINE, and Scopus library databases was conducted to find relevant articles. RESULTS The combination of radionuclide, chelator and required labeling chemistry was shown to have a significant influence on the stability, binding affinity and internalization rate, off-target interaction with normal tissues and blood proteins, interaction with enzymes, activity uptake and retention in excretory organs and activity uptake in tumors of radiolabeled bombesin antagonistic analogues. CONCLUSION Labeling chemistry has a very strong impact on the biodistribution profile of GRPRtargeting peptide based imaging probes and needs to be considered when designing a targeting probe for high contrast molecular imaging. Taking into account the complexity of in vivo interactions, it is not currently possible to accurately predict the optimal labeling approach. Therefore, a detailed in vivo characterization and optimization is essential for the rational design of imaging agents.
Collapse
Affiliation(s)
- Bogdan Mitran
- Department of Medicianl Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Faculty of Medicine, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicianl Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
54
|
Preclinical Evaluation of 99mTc-Labeled GRPR Antagonists maSSS/SES-PEG 2-RM26 for Imaging of Prostate Cancer. Pharmaceutics 2021; 13:pharmaceutics13020182. [PMID: 33573232 PMCID: PMC7912279 DOI: 10.3390/pharmaceutics13020182] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/17/2021] [Accepted: 01/22/2021] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Gastrin-releasing peptide receptor (GRPR) is an important target for imaging of prostate cancer. The wide availability of single-photon emission computed tomography/computed tomography (SPECT/CT) and the generator-produced 99mTc can be utilized to facilitate the use of GRPR-targeting radiotracers for diagnostics of prostate cancers. METHODS Synthetically produced mercaptoacetyl-Ser-Ser-Ser (maSSS)-PEG2-RM26 and mercaptoacetyl-Ser-Glu-Ser (maSES)-PEG2-RM26 (RM26 = d-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2) were radiolabeled with 99mTc and characterized in vitro using PC-3 cells and in vivo, using NMRI or PC-3 tumor bearing mice. SPECT/CT imaging and dosimetry calculations were performed for [99mTc]Tc-maSSS-PEG2-RM26. RESULTS Peptides were radiolabeled with high yields (>98%), demonstrating GRPR specific binding and slow internalization in PC-3 cells. [99mTc]Tc-maSSS-PEG2-RM26 outperformed [99mTc]Tc-maSES-PEG2-RM26 in terms of GRPR affinity, with a lower dissociation constant (61 pM vs 849 pM) and demonstrating higher tumor uptake. [99mTc]Tc-maSSS-PEG2-RM26 had tumor-to-blood, tumor-to-muscle, and tumor-to-bone ratios of 97 ± 56, 188 ± 32, and 177 ± 79, respectively. SPECT/CT images of [99mTc]Tc-maSSS-PEG2-RM26 clearly visualized the GRPR-overexpressing tumors. The dosimetry estimated for [99mTc]Tc-maSSS-PEG2-RM26 showed the highest absorbed dose in the small intestine (1.65 × 10-3 mGy/MBq), and the effective dose is 3.49 × 10-3 mSv/MBq. CONCLUSION The GRPR antagonist maSSS-PEG2-RM26 is a promising GRPR-targeting agent that can be radiolabeled through a single-step with the generator-produced 99mTc and used for imaging of GRPR-expressing prostate cancer.
Collapse
|
55
|
Makris G, Shegani A, Kankanamalage PHA, Kuchuk M, Bandari RP, Smith CJ, Hennkens HM. Preclinical Evaluation of Novel 64Cu-Labeled Gastrin-Releasing Peptide Receptor Bioconjugates for PET Imaging of Prostate Cancer. Bioconjug Chem 2021; 32:1290-1297. [PMID: 33434428 DOI: 10.1021/acs.bioconjchem.0c00656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report herein the preclinical evaluation of new [64Cu]Cu-gastrin-releasing peptide receptor (GRPR)-targeting tracers, employing the potent peptide antagonist DPhe-Gln-Trp-Ala-VaI-Gly-His-Sta-Leu-NH2 conjugated to NOTA (in 1) or NODAGA (in 2) chelators via a 6-aminohexanoic acid linker. The Cu-1/2 metalated peptides were synthesized by reacting 1/2 with CuCl2 and were characterized by LC-ESI-MS and HR-ESI-MS. Cu-1/2 exhibited high GRPR-binding affinities with IC50 values <3 nM, as measured in a competition assay using the GRPR-expressing human PC-3 prostate cancer cell line and [125I]I-Tyr4-BBN as the competing ligand. Tracers [64Cu]Cu-1/2 were prepared in quantitative radiochemical yield (by radio-HPLC), and their identities were confirmed by coelution with their Cu-1/2 standards via comparative HPLC studies. Lipophilicity was measured in 1-octanol/PBS (pH 7.4), and the negative log D7.4 values (≤-1) confirmed the anticipated hydrophilic character for [64Cu]Cu-1/2. Both tracers demonstrated excellent in vitro stability, with ≥98% remaining intact through 24 h at physiological conditions (PBS, pH 7.4, 37 °C). Biodistribution in PC-3 tumor-bearing mice demonstrated good tumor uptake (%ID/g at 4 h: 4.34 ± 0.71 for [64Cu]Cu-1, 3.92 ± 1.03 for [64Cu]Cu-2) and rapid renal clearance (≥87% ID at 4 h). Tumor uptake was receptor-mediated, as verified by parallel GRPR-blocking studies. Small-animal PET/CT imaging studies validated the biodistribution data. These preclinical data support that the [64Cu]Cu-1/2 tracers show promise for further development as diagnostic PET imaging agents of GRPR-expressing tumors.
Collapse
Affiliation(s)
- George Makris
- Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Antonio Shegani
- Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States.,Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research "Demokritos", 15310 Athens, Greece
| | | | - Marina Kuchuk
- Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Rajendra P Bandari
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65201, United States
| | - Charles J Smith
- Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65201, United States.,Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Heather M Hennkens
- Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States.,Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
56
|
Maina T, Thakur M. SPECT Radiochemistry. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00023-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
57
|
Aloj L, Attili B, Lau D, Caraco C, Lechermann LM, Mendichovszky IA, Harper I, Cheow H, Casey RT, Sala E, Gilbert FJ, Gallagher FA. The emerging role of cell surface receptor and protein binding radiopharmaceuticals in cancer diagnostics and therapy. Nucl Med Biol 2021; 92:53-64. [PMID: 32563612 DOI: 10.1016/j.nucmedbio.2020.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022]
Abstract
Targeting specific cell membrane markers for both diagnostic imaging and radionuclide therapy is a rapidly evolving field in cancer research. Some of these applications have now found a role in routine clinical practice and have been shown to have a significant impact on patient management. Several molecular targets are being investigated in ongoing clinical trials and show promise for future implementation. Advancements in molecular biology have facilitated the identification of new cancer-specific targets for radiopharmaceutical development.
Collapse
Affiliation(s)
- Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom.
| | - Bala Attili
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Doreen Lau
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Corradina Caraco
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Laura M Lechermann
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Iosif A Mendichovszky
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Ines Harper
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Heok Cheow
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ruth T Casey
- Department of Endocrinology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Evis Sala
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Fiona J Gilbert
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| |
Collapse
|
58
|
Preclinical Evaluation of the Copper-64 Labeled GRPR-Antagonist RM26 in Comparison with the Cobalt-55 Labeled Counterpart for PET-Imaging of Prostate Cancer. Molecules 2020; 25:molecules25245993. [PMID: 33352838 PMCID: PMC7766840 DOI: 10.3390/molecules25245993] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 01/21/2023] Open
Abstract
Gastrin-releasing peptide receptor (GRPR) is overexpressed in the majority of prostate cancers. This study aimed to investigate the potential of 64Cu (radionuclide for late time-point PET-imaging) for imaging of GRPR expression using NOTA-PEG2-RM26 and NODAGA-PEG2-RM26. Methods: NOTA/NODAGA-PEG2-RM26 were labeled with 64Cu and evaluated in GRPR-expressing PC-3 cells. Biodistribution of [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 was studied in PC-3 xenografted mice and compared to the biodistribution of [57Co]Co-NOTA/NODAGA-PEG2-RM26 at 3 and 24 h p.i. Preclinical PET/CT imaging was performed in tumor-bearing mice. NOTA/NODAGA-PEG2-RM26 were stably labeled with 64Cu with quantitative yields. In vitro, binding of [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 was rapid and GRPR-specific with slow internalization. In vivo, [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 bound specifically to GRPR-expressing tumors with fast clearance from blood and normal organs and displayed generally comparable biodistribution profiles to [57Co]Co-NOTA/NODAGA-PEG2-RM26; tumor uptake exceeded normal tissue uptake 3 h p.i.. Tumor-to-organ ratios did not increase significantly with time. [64Cu]Cu-NOTA-PEG2-RM26 had a significantly higher liver and pancreas uptake compared to other agents. 57Co-labeled radioconjugates showed overall higher tumor-to-non-tumor ratios, compared to the 64Cu-labeled counterparts. [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 was able to visualize GRPR-expression in a murine PC model using PET. However, [55/57Co]Co-NOTA/NODAGA-PEG2-RM26 provided better in vivo stability and overall higher tumor-to-non-tumor ratios compared with the 64Cu-labeled conjugates.
Collapse
|
59
|
Hübner R, Cheng X, Wängler B, Wängler C. Functional Hybrid Molecules for the Visualization of Cancer: PESIN-Homodimers Combined with Multimodal Molecular Imaging Probes for Positron Emission Tomography and Optical Imaging: Suited for Tracking of GRPR-Positive Malignant Tissue*. Chemistry 2020; 26:16349-16356. [PMID: 32618007 PMCID: PMC7756681 DOI: 10.1002/chem.202002386] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Indexed: 12/16/2022]
Abstract
We describe multimodal imaging probes for gastrin-releasing peptide receptor (GRPR)-specific targeting suited for positron emission tomography and optical imaging (PET/OI), consisting of PESIN (PEG3 -BBN7-14 ) dimers connected to multimodal imaging subunits. These multimodal agents comprise a fluorescent dye for OI and the chelator ((1,4,7-triazacyclononane-4,7-diyl)diacetic acid-1-glutaric acid) (NODA-GA) for PET radiometal isotope labelling. Special focus was put on the influence of the used dyes on the properties of the whole bioconjugates. For this, several compounds with different fluorescent dyes and non-dye carrying subunits were synthesized and investigated. As fluorescent dyes, dansyl, NBD, derivatives of fluorescein, coumarin and rhodamine as well as three pyrilium-based dyes were employed. Considerable influence of the charge of the colored unit on hydrophilicity as well as in vitro target receptor binding was observed and classified. High radiochemical yields and purities were found during radiolabeling of the multimodal imaging subunits as well as their GRPR-specific bioconjugates with 68 Ga. Examinations of the photophysical properties of both molecule species displayed no loss or alteration of fluorescence characteristics.
Collapse
Affiliation(s)
- Ralph Hübner
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| | - Xia Cheng
- Molecular Imaging and RadiochemistryDepartment of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| | - Björn Wängler
- Molecular Imaging and RadiochemistryDepartment of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| |
Collapse
|
60
|
Hofstetter M, Moon ES, D'Angelo F, Geissbühler L, Alberts I, Afshar-Oromieh A, Rösch F, Rominger A, Gourni E. Effect of the versatile bifunctional chelator AAZTA 5 on the radiometal labelling properties and the in vitro performance of a gastrin releasing peptide receptor antagonist. EJNMMI Radiopharm Chem 2020; 5:29. [PMID: 33258012 PMCID: PMC7704979 DOI: 10.1186/s41181-020-00115-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Background Gastrin Releasing Peptide receptor (GRPr)-based radioligands have shown great promise for diagnostic imaging of GRPr-positive cancers, such as prostate and breast. The present study aims at developing and evaluating a versatile GRPr-based probe for both PET/SPECT imaging as well as intraoperative and therapeutic applications. The influence of the versatile chelator AAZTA5 on the radiometal labelling properties and the in vitro performance of the generated radiotracers were thoroughly investigated. The GRPr-based antagonist D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 was functionalized with the chelator 6-[Bis (carboxymethyl)amino]-1,4-bis (carboyxmethyl)-6-methyl-1,4-diazepane (AAZTA5) through the spacer 4-amino-1-carboxymethyl-piperidine (Pip) to obtain AAZTA5-Pip-D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 (LF1). LF1 was radiolabelled with gallium-68 (PET), indium-111 (SPECT, intraoperative applications) and lutetium-177 (therapy, SPECT). In vitro evaluation included stability studies, determination of lipophilicity, protein-binding studies, determination of Kd and Bmax as well as internalization studies using the epithelial human prostate cancer cell line PC3. In vitro monotherapy as well as combination therapy studies were further performed to assess its applicability as a theranostic compound. Results LF1 was labelled with gallium-68, indium-111 and lutetium-177 within 5 min at room temperature (RT). The apparent molar activities (Am) were ranging between 50 and 60 GBq/μmol for the 68Ga-labelled LF1, 10–20 GBq/μmol for the 111In- and 177Lu-labelled LF1. The radiotracers were stable for a period of 4 h post labeling exhibiting a hydrophilic profile with an average of a LogDoctanol/PBS of − 3, while the bound activity to the human serum protein was approximately 10%. 68/natGa-LF1, 177/natLu-LF1 and 111/natIn-LF1 exhibited high affinity for the PC3 cells, with Kd values of 16.3 ± 2.4 nM, 10.3 ± 2.73 nM and 5.2 ± 1.9 nM, respectively, and the required concentration of the radiotracers to saturate the receptors (Bmax) was between 0.5 and 0.8 nM which corresponds to approximately 4 × 105 receptors per cell. Low specific internalization rate was found in cell culture, while the total specific cell surface bound uptake always exceeded the internalized activity. In vitro therapy studies showed that inhibition of PC3 cells growth is somewhat more efficient when combination of 177Lu-labelled LF1 with rapamycin is applied compared to 177Lu-laballed LF1 alone. Conclusion Encouraged by these promising in vitro data, preclinical evaluation of the LF1 precursor are planned in tumour models in vivo.
Collapse
Affiliation(s)
- Michael Hofstetter
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Euy Sung Moon
- Department of Chemistry - TRIGA site, Johannes Gutenberg - University Mainz, Mainz, Germany
| | - Fabio D'Angelo
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Lucien Geissbühler
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Ian Alberts
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Frank Rösch
- Department of Chemistry - TRIGA site, Johannes Gutenberg - University Mainz, Mainz, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Eleni Gourni
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, Freiburgstr. 18, 3010, Bern, Switzerland.
| |
Collapse
|
61
|
Sgouros G, Bodei L, McDevitt MR, Nedrow JR. Radiopharmaceutical therapy in cancer: clinical advances and challenges. Nat Rev Drug Discov 2020; 19:589-608. [PMID: 32728208 PMCID: PMC7390460 DOI: 10.1038/s41573-020-0073-9] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2020] [Indexed: 12/25/2022]
Abstract
Radiopharmaceutical therapy (RPT) is emerging as a safe and effective targeted approach to treating many types of cancer. In RPT, radiation is systemically or locally delivered using pharmaceuticals that either bind preferentially to cancer cells or accumulate by physiological mechanisms. Almost all radionuclides used in RPT emit photons that can be imaged, enabling non-invasive visualization of the biodistribution of the therapeutic agent. Compared with almost all other systemic cancer treatment options, RPT has shown efficacy with minimal toxicity. With the recent FDA approval of several RPT agents, the remarkable potential of this treatment is now being recognized. This Review covers the fundamental properties, clinical development and associated challenges of RPT.
Collapse
Affiliation(s)
- George Sgouros
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jessie R Nedrow
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
62
|
Radiochemical Synthesis and Evaluation of Novel Radioconjugates of Neurokinin 1 Receptor Antagonist Aprepitant Dedicated for NK1R-Positive Tumors. Molecules 2020; 25:molecules25163756. [PMID: 32824729 PMCID: PMC7466001 DOI: 10.3390/molecules25163756] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/18/2022] Open
Abstract
Aprepitant, a lipophilic and small molecular representative of neurokinin 1 receptor antagonists, is known for its anti-proliferative activity on numerous cancer cell lines that are sensitive to Substance P mitogen action. In the presented research, we developed two novel structural modifications of aprepitant to create aprepitant conjugates with different radionuclide chelators. All of them were radiolabeled with 68Ga and 177Lu radionuclides and evaluated in terms of their lipophilicity and stability in human serum. Furthermore, fully stable conjugates were examined in molecular modelling with a human neurokinin 1 receptor structure and in a competitive radioligand binding assay using rat brain homogenates in comparison to the aprepitant molecule. This initial research is in the conceptual stage to give potential theranostic-like radiopharmaceutical pairs for the imaging and therapy of neurokinin 1 receptor-overexpressing cancers.
Collapse
|
63
|
[ 99mTc]Tc-DB1 Mimics with Different-Length PEG Spacers: Preclinical Comparison in GRPR-Positive Models. Molecules 2020; 25:molecules25153418. [PMID: 32731473 PMCID: PMC7435657 DOI: 10.3390/molecules25153418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 02/07/2023] Open
Abstract
Background: The frequent overexpression of gastrin-releasing peptide receptors (GRPRs) in human cancers provides the rationale for delivering clinically useful radionuclides to tumor sites using peptide carriers. Radiolabeled GRPR antagonists, besides being safer for human use, have often shown higher tumor uptake and faster background clearance than agonists. We herein compared the biological profiles of the GRPR-antagonist-based radiotracers [99mTc]Tc-[N4-PEGx-DPhe6,Leu-NHEt13]BBN(6-13) (N4: 6-(carboxy)-1,4,8,11-tetraazaundecane; PEG: polyethyleneglycol): (i) [99mTc]Tc-DB7 (x = 2), (ii) [99mTc]Tc-DB13 (x = 3), and (iii) [99mTc]Tc-DB14 (x = 4), in GRPR-positive cells and animal models. The impact of in situ neprilysin (NEP)-inhibition on in vivo stability and tumor uptake was also assessed by treatment of mice with phosphoramidon (PA). Methods: The GRPR affinity of DB7/DB13/DB14 was determined in PC-3 cell membranes, and cell binding of the respective [99mTc]Tc-radioligands was assessed in PC-3 cells. Each of [99mTc]Tc-DB7, [99mTc]Tc-DB13, and [99mTc]Tc-DB14 was injected into mice without or with PA coinjection and 5 min blood samples were analyzed by HPLC. Biodistribution was conducted at 4 h postinjection (pi) in severe combined immunodeficiency disease (SCID) mice bearing PC-3 xenografts without or with PA coinjection. Results: DB7, -13, and -14 displayed single-digit nanomolar affinities for GRPR. The uptake rates of [99mTc]Tc-DB7, [99mTc]Tc-DB13, and [99mTc]Tc-DB14 in PC-3 cells was comparable and consistent with a radioantagonist profile. The radiotracers were found to be ≈70% intact in mouse blood and >94% intact after coinjection of PA. Treatment of mice with PA enhanced tumor uptake. Conclusions: The present study showed that increase of PEG-spacer length in the [99mTc]Tc-DB7-[99mTc]Tc-DB13-[99mTc]Tc-DB14 series had little effect on GRPR affinity, specific uptake in PC-3 cells, in vivo stability, or tumor uptake. A significant change in in vivo stability and tumor uptake was observed only after treatment of mice with PA, without compromising the favorably low background radioactivity levels.
Collapse
|
64
|
Bordon KDCF, Cologna CT, Fornari-Baldo EC, Pinheiro-Júnior EL, Cerni FA, Amorim FG, Anjolette FAP, Cordeiro FA, Wiezel GA, Cardoso IA, Ferreira IG, de Oliveira IS, Boldrini-França J, Pucca MB, Baldo MA, Arantes EC. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front Pharmacol 2020; 11:1132. [PMID: 32848750 PMCID: PMC7396678 DOI: 10.3389/fphar.2020.01132] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Animal poisons and venoms are comprised of different classes of molecules displaying wide-ranging pharmacological activities. This review aims to provide an in-depth view of toxin-based compounds from terrestrial and marine organisms used as diagnostic tools, experimental molecules to validate postulated therapeutic targets, drug libraries, prototypes for the design of drugs, cosmeceuticals, and therapeutic agents. However, making these molecules applicable requires extensive preclinical trials, with some applications also demanding clinical trials, in order to validate their molecular target, mechanism of action, effective dose, potential adverse effects, as well as other fundamental parameters. Here we go through the pitfalls for a toxin-based potential therapeutic drug to become eligible for clinical trials and marketing. The manuscript also presents an overview of the current picture for several molecules from different animal venoms and poisons (such as those from amphibians, cone snails, hymenopterans, scorpions, sea anemones, snakes, spiders, tetraodontiformes, bats, and shrews) that have been used in clinical trials. Advances and perspectives on the therapeutic potential of molecules from other underexploited animals, such as caterpillars and ticks, are also reported. The challenges faced during the lengthy and costly preclinical and clinical studies and how to overcome these hindrances are also discussed for that drug candidates going to the bedside. It covers most of the drugs developed using toxins, the molecules that have failed and those that are currently in clinical trials. The article presents a detailed overview of toxins that have been used as therapeutic agents, including their discovery, formulation, dosage, indications, main adverse effects, and pregnancy and breastfeeding prescription warnings. Toxins in diagnosis, as well as cosmeceuticals and atypical therapies (bee venom and leech therapies) are also reported. The level of cumulative and detailed information provided in this review may help pharmacists, physicians, biotechnologists, pharmacologists, and scientists interested in toxinology, drug discovery, and development of toxin-based products.
Collapse
Affiliation(s)
- Karla de Castro Figueiredo Bordon
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Takeno Cologna
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ernesto Lopes Pinheiro-Júnior
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Gobbi Amorim
- Postgraduate Program in Pharmaceutical Sciences, Vila Velha University, Vila Velha, Brazil
| | | | - Francielle Almeida Cordeiro
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gisele Adriano Wiezel
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Iara Aimê Cardoso
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isabela Gobbo Ferreira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora Sousa de Oliveira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Mateus Amaral Baldo
- Health and Science Institute, Paulista University, São José do Rio Pardo, Brazil
| | - Eliane Candiani Arantes
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
65
|
Clinical application of Fluciclovine PET, choline PET and gastrin-releasing polypeptide receptor (bombesin) targeting PET in prostate cancer. Curr Opin Urol 2020; 30:641-648. [PMID: 32701717 DOI: 10.1097/mou.0000000000000794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The aim of this review is to explore the clinical application of different PET radiopharmaceuticals in prostate cancer (PCa), beyond inhibitors of the prostate-specific membrane antigen (PSMA). RECENT FINDINGS Choline PET represented in the last decades the standard of reference for PET imaging in PCa and has been recently included in clinical trials evaluating the efficacy of metastasis-directed therapy in oligo-metastatic disease. Fluciclovine, as synthetic amino acid, has been proposed for investigating PCa. The results obtained by the first prospective studies led to FDA approval in 2016 in patients with biochemical recurrence. Recently, phase II/III trials explored its accuracy compared with PSMA PET and its impact on patient management. Imaging the gastrin-releasing polypeptide receptor (GRPR) recently drawn attention. Radio-labelled GRPR antagonists have the potential to be used as theranostic agents. Further evaluation is needed to understand the relation between GRPR expression and hormonal-resistant PCa, and for tumors characterized by heterogeneity of receptors expressed (e.g. PSMA-negative) on their cell surface. SUMMARY Other new generation PET tracers may play an important role in PCa, namely in case of PSMA-negative phenotypes.
Collapse
|
66
|
Mena E, Black PC, Rais-Bahrami S, Gorin M, Allaf M, Choyke P. Novel PET imaging methods for prostate cancer. World J Urol 2020; 39:687-699. [PMID: 32671604 DOI: 10.1007/s00345-020-03344-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Prostate cancer is a common neoplasm but conventional imaging methods such as CT and bone scan are often insensitive. A new class of PET agents have emerged to diagnose and manage prostate cancer. METHODS The relevant literature on PET imaging agents for prostate cancer was reviewed. RESULTS This review shows a broad range of PET imaging agents, the most successful of which is prostate specific membrane antigen (PSMA) PET. Other agents either lack the sensitivity or specificity of PSMA PET. CONCLUSION Among the available PET agents for prostate cancer, PSMA PET has emerged as the leader. It is likely to have great impact on the diagnosis, staging and management of prostate cancer patients.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Program, National Cancer Institute, 10 Center Dr, Bldg 10, Room B3B69F, Bethesda, MD, 20892-1088, USA
| | - Peter C Black
- University of British Columbia, Vancouver, BC, Canada
| | | | - Michael Gorin
- Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Mohamad Allaf
- Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter Choyke
- Molecular Imaging Program, National Cancer Institute, 10 Center Dr, Bldg 10, Room B3B69F, Bethesda, MD, 20892-1088, USA.
| |
Collapse
|
67
|
Hoppenz P, Els-Heindl S, Beck-Sickinger AG. Peptide-Drug Conjugates and Their Targets in Advanced Cancer Therapies. Front Chem 2020; 8:571. [PMID: 32733853 PMCID: PMC7359416 DOI: 10.3389/fchem.2020.00571] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer became recently the leading cause of death in industrialized countries. Even though standard treatments achieve significant effects in growth inhibition and tumor elimination, they cause severe side effects as most of the applied drugs exhibit only minor selectivity for the malignant tissue. Hence, specific addressing of tumor cells without affecting healthy tissue is currently a major desire in cancer therapy. Cell surface receptors, which bind peptides are frequently overexpressed on cancer cells and can therefore be considered as promising targets for selective tumor therapy. In this review, the benefits of peptides as tumor homing agents are presented and an overview of the most commonly addressed peptide receptors is given. A special focus was set on the bombesin receptor family and the neuropeptide Y receptor family. In the second part, the specific requirements of peptide-drug conjugates (PDC) and intelligent linker structures as an essential component of PDC are outlined. Furthermore, different drug cargos are presented including classical and recent toxic agents as well as radionuclides for diagnostic and therapeutic approaches. In the last part, boron neutron capture therapy as advanced targeted cancer therapy is introduced and past and recent developments are reviewed.
Collapse
Affiliation(s)
- Paul Hoppenz
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Sylvia Els-Heindl
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
68
|
Kaloudi A, Kanellopoulos P, Radolf T, Chepurny OG, Rouchota M, Loudos G, Andreae F, Holz GG, Nock BA, Maina T. [ 99mTc]Tc-DGA1, a Promising CCK 2R-Antagonist-Based Tracer for Tumor Diagnosis with Single-Photon Emission Computed Tomography. Mol Pharm 2020; 17:3116-3128. [PMID: 32568549 DOI: 10.1021/acs.molpharmaceut.0c00605] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Radiolabeled gastrin analogues have been proposed for theranostics of cholecystokinin subtype 2 receptor (CCK2R)-positive cancer. Peptide radioligands based on other receptor antagonists have displayed superior pharmacokinetics and higher biosafety than agonists. Here, we present DGA1, a derivative of the nonpeptidic CCK2R antagonist Z-360 carrying an acyclic tetraamine, for [99mTc]Tc labeling. Preclinical comparison of [99mTc]Tc-DGA1 with [99mTc]Tc-DG2 (CCK2R-agonist reference) was conducted in HEK293-CCK2R/CCK2i4svR cells and mice models, qualifying [99mTc]Tc-DGA1 for further study in patients with CCK2R-positive tumors and single-photon emission computed tomography/CT.
Collapse
Affiliation(s)
- Aikaterini Kaloudi
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece
| | | | - Thorsten Radolf
- piCHEM Forschungs-und Entwicklungs GmbH, Parkring 3, 8074 Grambach, Austria
| | - Oleg G Chepurny
- SUNY Upstate Medical University, IHP 4310 at 505 Irving Avenue, Syracuse, New York 13210, United States
| | - Maritina Rouchota
- BIOEMTECH, Lefkippos Attica Technology Park NCSR "Demokritos", 15310 Athens, Greece
| | - George Loudos
- BIOEMTECH, Lefkippos Attica Technology Park NCSR "Demokritos", 15310 Athens, Greece
| | - Fritz Andreae
- piCHEM Forschungs-und Entwicklungs GmbH, Parkring 3, 8074 Grambach, Austria
| | - George G Holz
- SUNY Upstate Medical University, IHP 4310 at 505 Irving Avenue, Syracuse, New York 13210, United States
| | | | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece
| |
Collapse
|
69
|
Song J, Huang S, Zhang Z, Jia B, Xie H, Kai M, Zhang W. SPA: a peptide antagonist that acts as a cell-penetrating peptide for drug delivery. Drug Deliv 2020; 27:91-99. [PMID: 31870182 PMCID: PMC6968712 DOI: 10.1080/10717544.2019.1706669] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Although cell-penetrating peptides (CPPs) has been proven to be efficient transporter for drug delivery, ideal peptide vectors for tumor therapy are still being urgently sought. Peptide antagonists have attracted substantial attention as targeting molecules because of their high tumor accumulation and antitumor activity compared with agonists. SPA, a derivative of substance P, is a potent antagonist that exhibits antitumor activity. Based on the amino acid composition of SPA, we speculate that it can translocate across cell membranes as CPPs do. In this study, our results demonstrated that SPA could enter cells similarly to a CPP. As a vector, SPA could efficiently deliver camptothecin and plasmids into cells. In addition, our results showed that SPA exhibited low toxicity to normal cells and high enzymatic stability. Taken together, our results validated the ability of SPA for efficient drug delivery. More importantly, our study opens a new avenue for designing ideal CPPs based on peptide antagonists.
Collapse
Affiliation(s)
- Jingjing Song
- Institute of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Sujie Huang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Zhengzheng Zhang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bo Jia
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Huan Xie
- Institute of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ming Kai
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Wei Zhang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
70
|
Structural modifications of amino acid sequences of radiolabeled peptides for targeted tumor imaging. Bioorg Chem 2020; 99:103802. [DOI: 10.1016/j.bioorg.2020.103802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022]
|
71
|
Gruber L, Jiménez-Franco LD, Decristoforo C, Uprimny C, Glatting G, Hohenberger P, Schoenberg SO, Reindl W, Orlandi F, Mariani M, Jaschke W, Virgolini I. MITIGATE-NeoBOMB1, a Phase I/IIa Study to Evaluate Safety, Pharmacokinetics, and Preliminary Imaging of 68Ga-NeoBOMB1, a Gastrin-Releasing Peptide Receptor Antagonist, in GIST Patients. J Nucl Med 2020; 61:1749-1755. [PMID: 32332143 DOI: 10.2967/jnumed.119.238808] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/28/2020] [Indexed: 12/13/2022] Open
Abstract
Gastrin-releasing peptide receptors (GRPRs) are potential molecular imaging targets in a variety of tumors. Recently, a 68Ga-labeled antagonist to GRPRs, NeoBOMB1, was developed for PET. We report on the outcome of a phase I/IIa clinical trial (EudraCT 2016-002053-38) within the EU-FP7 project Closed-loop Molecular Environment for Minimally Invasive Treatment of Patients with Metastatic Gastrointestinal Stromal Tumors ('MITIGATE') (grant agreement no. 602306) in patients with oligometastatic gastrointestinal stromal tumors (GIST). Methods: The main objectives were evaluation of safety, biodistribution, dosimetry, and preliminary tumor targeting of 68Ga-NeoBOMB1 in patients with advanced tyrosine-kinase inhibitors-treated GIST using PET/CT. Six patients with histologically confirmed GIST and unresectable primary lesion or metastases undergoing an extended protocol for detailed pharmacokinetic analysis were included. 68Ga-NeoBOMB1 was prepared using a kit procedure with a licensed 68Ge/68Ga generator. 68Ga-NeoBOMB1 (3 MBq/kg of body weight) was injected intravenously, and safety parameters were assessed. PET/CT included dynamic imaging at 5, 11, and 19 min as well as static imaging at 1, 2, and 3-4 h after injection for dosimetry calculations. Venous blood samples and urine were collected for pharmacokinetic analysis. Tumor targeting was assessed on a per-lesion and per-patient basis. Results: 68Ga-NeoBOMB1 (50 μg) was prepared with high radiochemical purity (yield > 97%). Patients received 174 ± 28 MBq of the radiotracer, which was well tolerated in all patients over a follow-up period of 4 wk. Dosimetry calculations revealed a mean effective dose of 0.029 ± 0.06 mSv/MBq, with the highest organ dose to the pancreas (0.274 ± 0.099 mSv/MBq). Mean plasma half-life was 27.3 min with primarily renal clearance (mean 25.7% ± 5.4% of injected dose 4 h after injection). Plasma metabolite analyses revealed high stability; metabolites were detected only in the urine. In 3 patients, a significant uptake with increasing maximum SUVs (SUVmax at 2 h after injection: 4.3-25.9) over time was found in tumor lesions. Conclusion: This phase I/IIa study provides safety data for 68Ga-NeoBOMB1, a promising radiopharmaceutical for targeting GRPR-expressing tumors. Safety profiles and pharmacokinetics are suitable for PET imaging, and absorbed dose estimates are comparable to those of other 68Ga-labeled radiopharmaceuticals used in clinical routine.
Collapse
Affiliation(s)
- Leonhard Gruber
- Department of Radiology, Medical University Innsbruck, Innsbruck, Austria
| | - Luis David Jiménez-Franco
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Uprimny
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan O Schoenberg
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang Reindl
- Klinikum Mannheim II, Medizinische Klinik, Mannheim, Germany; and
| | - Francesca Orlandi
- Advanced Accelerator Applications, a Novartis Company, Colleretto Giacosa TO, Italy
| | - Maurizio Mariani
- Advanced Accelerator Applications, a Novartis Company, Colleretto Giacosa TO, Italy
| | - Werner Jaschke
- Department of Radiology, Medical University Innsbruck, Innsbruck, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
72
|
Baratto L, Duan H, Mäcke H, Iagaru A. Imaging the Distribution of Gastrin-Releasing Peptide Receptors in Cancer. J Nucl Med 2020; 61:792-798. [DOI: 10.2967/jnumed.119.234971] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/06/2020] [Indexed: 01/01/2023] Open
|
73
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
74
|
Tu Y, Tao J, Wang F, Liu P, Han Z, Li Z, Ma Y, Gu Y. A novel peptide targeting gastrin releasing peptide receptor for pancreatic neoplasm detection. Biomater Sci 2020; 8:2682-2693. [DOI: 10.1039/d0bm00162g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The designed novel peptide GB-6 with targeted GRPR-binding possesses more favorable pharmacokinetic properties and metabolic stability, as well as superior tumor-targeting ability in pancreatic cancer models, relative to BBN7–14.
Collapse
Affiliation(s)
- Yuanbiao Tu
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Ji Tao
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Fang Wang
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Peifei Liu
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Zhihao Han
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Zhaolun Li
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Yi Ma
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| | - Yueqing Gu
- State Key Laboratory of Natural Medicine
- Department of Biomedical Engineering
- School of Engineering
- China Pharmaceutical University
- Nanjing 210009
| |
Collapse
|
75
|
Hoppenz P, Els‐Heindl S, Beck‐Sickinger AG. Identification and stabilization of a highly selective gastrin‐releasing peptide receptor agonist. J Pept Sci 2019; 25:e3224. [DOI: 10.1002/psc.3224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Paul Hoppenz
- Institute of BiochemistryLeipzig University Leipzig Germany
| | | | | |
Collapse
|
76
|
Mitran B, Thisgaard H, Rinne S, Dam JH, Azami F, Tolmachev V, Orlova A, Rosenström U. Selection of an optimal macrocyclic chelator improves the imaging of prostate cancer using cobalt-labeled GRPR antagonist RM26. Sci Rep 2019; 9:17086. [PMID: 31745219 PMCID: PMC6863848 DOI: 10.1038/s41598-019-52914-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022] Open
Abstract
Gastrin-releasing peptide receptors (GRPRs) are promising targets in oligometastatic prostate cancer. We have recently used 55Co (T1/2 = 17.5 h) as a label for next day PET imaging of GRPR expression obtaining high imaging contrast. The radionuclide-chelator combination can significantly influence the biodistribution of radiopeptides. Therefore, in this study, we hypothesized that the properties of 55Co-labeled PEG2-RM26 can be improved by identifying the optimal macrocyclic chelator. All analogues (X-PEG2-RM26, X = NOTA,NODAGA,DOTA,DOTAGA) were successfully labeled with radiocobalt with high yields and demonstrated high stability. The radiopeptides bound specifically and with picomolar affinity to GRPR and their cellular processing was characterized by low internalization. The best binding capacity was found for DOTA-PEG2-RM26. Ex vivo biodistribution in PC-3 xenografted mice was characterized by rapid blood clearance via renal excretion. Tumor uptake was similar for all conjugates at 3 h pi, exceeding the uptake in all other organs. Higher kidney uptake and longer retention were associated with N-terminal negative charge (DOTAGA-containing conjugate). Tumor-to-organ ratios increased over time for all constructs, although significant chelator-dependent differences were observed. Concordant with affinity measurements, DOTA-analog had the best retention of activity in tumors, resulting in the highest tumor-to-blood ratio 24 h pi, which translated into high contrast PET/CT imaging (using 55Co).
Collapse
Affiliation(s)
- Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Helge Thisgaard
- PET & Cyclotron Unit, Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sara Rinne
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Johan Hygum Dam
- PET & Cyclotron Unit, Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Frishta Azami
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
77
|
Zhang W, Fan W, Ottemann BM, Alshehri S, Garrison JC. Development of Improved Tumor-Residualizing, GRPR-Targeted Agents: Preclinical Comparison of an Endolysosomal Trapping Approach in Agonistic and Antagonistic Constructs. J Nucl Med 2019; 61:443-450. [PMID: 31601697 DOI: 10.2967/jnumed.119.231282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
Receptor-targeted radiopharmaceuticals based on low-molecular-weight carriers offer many clinically advantageous attributes relative to macromolecules but have generally been hampered by their rapid clearance from tumors, thus diminishing tumor-to-nontarget tissue ratios. Herein, we present a strategy using irreversible inhibitors (E-64 derivative) of cysteine cathepsins (CCs) as trapping agents to increase the tumor retention of receptor-targeted agents. Methods: We incorporated these CC-trapping agents into agonistic and antagonistic pharmacophores targeting the gastrin-releasing peptide receptor (GRPR). The synthesized radioconjugates with either an incorporated CC inhibitor or a matching control were examined using in vitro and in vivo models of the GRPR-positive, PC-3 human prostate cancer cell line. Results: From the in vitro studies, multiple techniques confirmed that the CC-trapping, GRPR-targeted constructs were able to increase cellular retention by forming intracellular macromolecule adducts. In PC-3 tumor-bearing xenograft mice, the CC-trapping, GRPR-targeted agonistic and antagonistic constructs led to an approximately 2-fold increase in tumor retention with a corresponding improvement in most tumor-to-nontarget tissue ratios over 72 h. Conclusion: CC endolysosomal trapping provides a pathway to increase the efficacy and clinical potential of low-molecular-weight, receptor-targeted agents.
Collapse
Affiliation(s)
- Wenting Zhang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska.,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Wei Fan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska.,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sameer Alshehri
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska.,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jered C Garrison
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska .,Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska; and.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
78
|
Pooja D, Gunukula A, Gupta N, Adams DJ, Kulhari H. Bombesin receptors as potential targets for anticancer drug delivery and imaging. Int J Biochem Cell Biol 2019; 114:105567. [DOI: 10.1016/j.biocel.2019.105567] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022]
|
79
|
Stability Evaluation and Stabilization of a Gastrin-Releasing Peptide Receptor (GRPR) Targeting Imaging Pharmaceutical. Molecules 2019; 24:molecules24162878. [PMID: 31398865 PMCID: PMC6720803 DOI: 10.3390/molecules24162878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/24/2019] [Accepted: 08/06/2019] [Indexed: 01/15/2023] Open
Abstract
The prostate-specific membrane antigen (PSMA) and gastrin-releasing peptide receptor (GRPR) are identified as important targets on prostate cancer. Receptor-targeting radiolabeled imaging pharmaceuticals with high affinity and specificity are useful in studying and monitoring biological processes and responses. Two potential imaging pharmaceuticals, AMBA agonist (where AMBA = DO3A-CH2CO-G-[4-aminobenzyl]- Gln-Trp-Ala-Val-Gly-His-Leu-Met-NH2) and RM1 antagonist (where RM1 = DO3A-CH2CO-G-[4-aminobenzyl]-D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2), have demonstrated high binding affinity (IC50) to GRP receptors and high tumor uptake. Antagonists, despite the poor tumor cell internalization properties, can show clearer images and pharmacokinetic profiles by virtue of their higher tumor uptake in animal models compared to agonists. For characterization, development, and translation of a potential imaging pharmaceutical into the clinic, it must be evaluated in a series of tests, including in vitro cell binding assays, in vitro buffer and serum stability studies, the biodistribution of the radiolabeled material, and finally imaging studies in preclinical animal models. Data related to acetate buffer, mouse, canine, and human sera stability of 177Lu-labeled RM1 are presented here and compared with the acetate buffer and sera stability data of AMBA agonist. The samples of 177Lu-labeled RM1 with a high radioconcentration degrade faster than low-radioconcentration samples upon storage at 2–8 °C. Addition of stabilizers, ascorbic acid and gentisic acid, improve the stability of 177Lu-labeled RM1 significantly with gentisic acid being more efficient than ascorbic acid as a stabilizer. The degradation kinetics of 177Lu-labeled AMBA and RM1 in sera follow the order (fastest to slowest): mouse > canine > human sera. Finally, 177Lu-labeled RM1 antagonist is slower to degrade in mouse, canine, and human sera than 177Lu-labeled AMBA agonist, further suggesting that an antagonist is a more promising candidate than agonist for the positron emission tomography (PET) imaging and therapy of prostate cancer patients.
Collapse
|
80
|
Oroujeni M, Abouzayed A, Lundmark F, Mitran B, Orlova A, Tolmachev V, Rosenström U. Evaluation of Tumor-Targeting Properties of an Antagonistic Bombesin Analogue RM26 Conjugated with a Non-Residualizing Radioiodine Label Comparison with a Radiometal-Labelled Counterpart. Pharmaceutics 2019; 11:pharmaceutics11080380. [PMID: 31382362 PMCID: PMC6724035 DOI: 10.3390/pharmaceutics11080380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
Radiolabelled antagonistic bombesin analogues are successfully used for targeting of gastrin-releasing peptide receptors (GRPR) that are overexpressed in prostate cancer. Internalization of antagonistic bombesin analogues is slow. We hypothesized that the use of a non-residualizing radioiodine label might not affect the tumour uptake but would reduce the retention in normal organs, where radiopharmaceutical would be internalized. To test this hypothesis, tyrosine was conjugated via diethylene glycol linker to N-terminus of an antagonistic bombesin analogue RM26 to form Tyr-PEG2-RM26. [111In]In-DOTA-PEG2-RM26 was used as a control with a residualizing label. Tyr-PEG2-RM26 was labelled with 125I with 95% radiochemical purity and retained binding specificity to GRPR. The IC50 values for Tyr-PEG2-RM26 and DOTA-PEG2-RM26 were 1.7 ± 0.3 nM and 3.3 ± 0.5 nM, respectively. The cellular processing of [125I]I-Tyr-PEG2-RM26 by PC-3 cells showed unusually fast internalization. Biodistribution showed that uptake in pancreas and tumour was GRPR-specific for both radioconjugates. Blood clearance of [125I]I-Tyr-PEG2-RM26 was appreciably slower and activity accumulation in all organs was significantly higher than for [111In]In-DOTA-PEG2-RM26. Tumor uptake of [111In]In-DOTA-PEG2-RM26 was significantly higher than for [125I]I-Tyr-PEG2-RM26, resulting in higher tumour-to-organ ratio for [111In]In-DOTA-PEG2-RM26 at studied time points. Incorporation of amino acids with hydrophilic side-chains next to tyrosine might overcome the problems associated with the use of tyrosine as a prosthetic group for radioiodination.
Collapse
Affiliation(s)
- Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Fanny Lundmark
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, SE-750 03 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden.
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
81
|
Farzipour S, Hosseinimehr SJ. Correlation between in vitro and in vivo Data of Radiolabeled Peptide for Tumor Targeting. Mini Rev Med Chem 2019; 19:950-960. [DOI: 10.2174/1389557519666190304120011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/28/2018] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
Tumor-targeting peptides have been generally developed for the overexpression of tumor specific receptors in cancer cells. The use of specific radiolabeled peptide allows tumor visualization by single photon emission computed tomography (SPECT) and positron emission tomography (PET) tools. The high affinity and specific binding of radiolabeled peptide are focusing on tumoral receptors. The character of the peptide itself, in particular, its complex molecular structure and behaviors influence on its specific interaction with receptors which are overexpressed in tumor. This review summarizes various strategies which are applied for the expansion of radiolabeled peptides for tumor targeting based on in vitro and in vivo specific tumor data and then their data were compared to find any correlation between these experiments. With a careful look at previous studies, it can be found that in vitro unblock-block ratio was unable to correlate the tumor to muscle ratio and the success of radiolabeled peptide for in vivo tumor targeting. The introduction of modifiers’ approaches, nature of peptides, and type of chelators and co-ligands have mixed effect on the in vitro and in vivo specificity of radiolabeled peptides.
Collapse
Affiliation(s)
- Soghra Farzipour
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
82
|
Liolios C, Sachpekidis C, Schäfer M, Kopka K. Bispecific radioligands targeting prostate-specific membrane antigen and gastrin-releasing peptide receptors on the surface of prostate cancer cells. J Labelled Comp Radiopharm 2019; 62:510-522. [DOI: 10.1002/jlcr.3749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/15/2019] [Accepted: 05/03/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Christos Liolios
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
- German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| |
Collapse
|
83
|
Zhang J, Singh A, Kulkarni HR, Schuchardt C, Müller D, Wester HJ, Maina T, Rösch F, van der Meulen NP, Müller C, Mäcke H, Baum RP. From Bench to Bedside-The Bad Berka Experience With First-in-Human Studies. Semin Nucl Med 2019; 49:422-437. [PMID: 31470935 DOI: 10.1053/j.semnuclmed.2019.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Precision oncology is being driven by rapid advances in novel diagnostics and therapeutic interventions, with treatments targeted to the needs of individual patients on the basis of genetic, biomarker, phenotypic, or psychosocial characteristics that distinguish a given patient from other patients with similar clinical presentations. Inherent in the theranostics paradigm is the assumption that diagnostic test results can precisely determine whether an individual is likely to benefit from a specific treatment. As part and integral in the current era of precision oncology, theranostics in the context of nuclear medicine aims to identify the appropriate molecular targets in neoplasms (diagnostic tool), so that the optimal ligands and radionuclides (therapeutic tool) with favorable labeling chemistry can be selected for personalized management of a specific disease, taking into consideration the specific patient, and subsequently monitor treatment response. Over the past two decades, the use of gallium-68 labeled peptides for somatostatin receptor (SSTR)-targeted PET/CT (or PET/MRI) imaging followed by lutetium-177 and yttrium-90 labeled SSTR-agonist for peptide receptor radionuclide therapy has demonstrated remarkable success in the management of neuroendocrine neoplasms, and paved the way to other indications of theranostics. Rapid advances are being made in the development of other peptide-based radiopharmaceuticals, small molecular-weight ligands and with newer radioisotopes with more favorable kinetics, potentially useful for theranostics strategies for the clinical application. The present review features the Bad Berka experience with first-in-human studies of new radiopharmaceuticals, for example, prostate-specific membrane antigen ligand, gastrin-releasing peptide receptor, neurotensin receptor 1 ligand, novel SSTR-targeting peptides and nonpeptide, and bone-seeking radiopharmaceuticals. Also new radioisotopes, for example, actinium (225Ac), copper (64Cu), scandium (44Sc), and terbium (152Tb/161Tb) will be discussed briefly demonstrating the development from basic science to precision oncology in the clinical setting.
Collapse
Affiliation(s)
- Jingjing Zhang
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Aviral Singh
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Harshad R Kulkarni
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Christiane Schuchardt
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Dirk Müller
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Hans-J Wester
- Institute for Radiopharmaceutical Chemistry, Technische Universität München, Garching, Germany
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", Athens, Greece
| | - Frank Rösch
- Institute of Nuclear Chemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland; (
- )Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| | - Helmut Mäcke
- Department of Nuclear Medicine, University Hospital of Freiburg, Freiburg, Germany
| | - Richard P Baum
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany.
| |
Collapse
|
84
|
Advances in the strategies for designing receptor-targeted molecular imaging probes for cancer research. J Control Release 2019; 305:1-17. [DOI: 10.1016/j.jconrel.2019.04.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/09/2019] [Accepted: 04/21/2019] [Indexed: 12/24/2022]
|
85
|
Lymperis E, Kaloudi A, Kanellopoulos P, Krenning EP, de Jong M, Maina T, Nock BA. Comparative evaluation of the new GRPR-antagonist 111 In-SB9 and 111 In-AMBA in prostate cancer models: Implications of in vivo stability. J Labelled Comp Radiopharm 2019; 62:646-655. [PMID: 30963606 DOI: 10.1002/jlcr.3733] [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] [Received: 12/02/2018] [Revised: 03/04/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022]
Abstract
Gastrin-releasing peptide receptors (GRPRs) are overexpressed in prostate cancer, representing attractive targets for diagnosis and therapy with bombesin (BBN)-like radioligands. GRPR-antagonists have lately attracted much attention owing to inherent biosafety and favorable pharmacokinetics. We herein present the GRPR-antagonist SB9 structurally resembling the known BBN-based agonist AMBA (SB9 = [Leu13 NHEt-desMet14 ]AMBA). The profiles of 111 In-SB9 and 111 In-AMBA were directly compared in PC-3 cells and tumor-bearing mice. SB9 and AMBA displayed high GRPR affinities. 111 In-AMBA strongly internalized in PC-3 cells, while 111 In-SB9 remained bound on the cell surface showing a typical GRPR-radioantagonist profile. 111 In-SB9 was more stable than 111 In-AMBA, but coinjection of the neprilysin (NEP) inhibitor phosphoramidon (PA) stabilized both in vivo. The radioligands displayed high tumor uptake (20.23 ± 3.41 %ID/g and 18.53 ± 1.54 %ID/g, respectively, at 4 hours pi), but 111 In-SB9 washed faster from background. PA coinjection led to significant increase of tumor uptake, combined with better clearance for 111 In-SB9. In short, this study has revealed superior pharmacokinetics and higher stability for the GRPR-antagonist 111 In-SB9 vs the corresponding agonist 111 In-AMBA consolidating previous evidence that GRPR antagonists are preferable to agonists for tumor imaging and therapy. It has also demonstrated that further pharmacokinetic improvements were feasible by in situ metabolic radioligand stabilization using PA.
Collapse
Affiliation(s)
| | | | | | - Eric P Krenning
- Cytrotron Rotterdam BV, Erasmus MC, Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", Athens, Greece
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", Athens, Greece
| |
Collapse
|
86
|
Oliveira MC, Correia JDG. Biomedical applications of radioiodinated peptides. Eur J Med Chem 2019; 179:56-77. [PMID: 31238251 DOI: 10.1016/j.ejmech.2019.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 02/08/2023]
Abstract
The overexpression of peptide receptors in certain tumors as compared to endogeneous expression levels represents the molecular basis for the design of peptide-based tools for targeted nuclear imaging and therapy. Receptor targeting with radiolabelled peptides became a very important imaging and/or therapeutic approach in nuclear medicine and oncology. A great variety of peptides has been radiolabelled with clinical relevant radionuclides, such as radiometals and radiohalogens. However, to the best of our knowledge concise and updated reviews providing information about the biomedical application of radioiodinated peptides are still missing. This review outlines the synthetic efforts in the preparation of radioiodinated peptides highlighting the importance of radioiodine in nuclear medicine, giving an overview of the most relevant radioiodination strategies that have been employed and describes relevant examples of their use in the biomedical field.
Collapse
Affiliation(s)
- Maria Cristina Oliveira
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal.
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal.
| |
Collapse
|
87
|
Ramos-Alvarez I, Lee L, Mantey SA, Jensen RT. Development and Characterization of a Novel, High-Affinity, Specific, Radiolabeled Ligand for BRS-3 Receptors. J Pharmacol Exp Ther 2019; 369:454-465. [PMID: 30971479 PMCID: PMC6519687 DOI: 10.1124/jpet.118.255141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/05/2019] [Indexed: 02/06/2023] Open
Abstract
Bombesin (Bn) receptor subtype 3(BRS-3) is an orphan G-protein-coupled receptor of the Bn family, which does not bind any natural Bn peptide with high affinity. Receptor knockout studies show that the animals develop diabetes, obesity, altered temperature control, and other central nervous system (CNS)/endocrine/gastrointestinal changes. It is present in CNS, peripheral tissues, and tumors; however, its role in normal physiology/pathophysiology, as well as its receptor localization/pharmacology is largely unknown, in part due to the lack of a convenient, specific, direct radiolabeled ligand. This study was designed to address this problem and to develop and characterize a specific radiolabeled ligand for BRS-3. The peptide antagonist Bantag-1 had >10,000-fold selectivity for human BRS-3 (hBRS-3) over other mammalian Bn receptors (BnRs) [i.e., gastrin-releasing peptide receptor (GRPR) and neuromedin B receptor (NMBR)]. Using iodogen and basic conditions, it was radiolabeled to high specific activity (2200 Ci/mmol) and found to bind with high affinity/specificity to hBRS-3. Binding was saturable, rapid, and reversible. The ligand only interacted with known BRS-3 ligands, and not with other specific GRPR/NMBR ligands or ligands for unrelated receptors. The magnitude of 125I-Bantag-1 binding correlated with BRS-3 mRNA expression and the magnitude of activation of phospholipase C in lung cancer cells, as well as readily identifying BRS-3 in lung cancer cells and normal tissues, allowing the direct assessment of BRS-3 receptor pharmacology/numbers on cells containing BRS-3 with other BnRs, which is usually the case. This circumvents the need for subtraction assays, which are now frequently used to assess BRS-3 indirectly using radiolabeled pan-ligands, which interact with all BnRs.
Collapse
Affiliation(s)
- Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Samuel A Mantey
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Robert T Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
88
|
Mitran B, Rinne SS, Konijnenberg MW, Maina T, Nock BA, Altai M, Vorobyeva A, Larhed M, Tolmachev V, de Jong M, Rosenström U, Orlova A. Trastuzumab cotreatment improves survival of mice with PC-3 prostate cancer xenografts treated with the GRPR antagonist 177 Lu-DOTAGA-PEG 2 -RM26. Int J Cancer 2019; 145:3347-3358. [PMID: 31077356 PMCID: PMC6852655 DOI: 10.1002/ijc.32401] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/19/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022]
Abstract
Gastrin‐releasing peptide receptors (GRPRs) are overexpressed in prostate cancer and are suitable for targeted radionuclide therapy (TRT). We optimized the bombesin‐derived GRPR‐antagonist PEG2‐RM26 for labeling with 177Lu and further determined the effect of treatment with 177Lu‐labeled peptide alone or in combination with the anti‐HER2 antibody trastuzumab in a murine model. The PEG2‐RM26 analog was coupled to NOTA, NODAGA, DOTA and DOTAGA chelators. The peptide‐chelator conjugates were labeled with 177Lu and characterized in vitro and in vivo. A preclinical therapeutic study was performed in PC‐3 xenografted mice. Mice were treated with intravenous injections (6 cycles) of (A) PBS, (B) DOTAGA‐PEG2‐RM26, (C) 177Lu‐DOTAGA‐PEG2‐RM26, (D) trastuzumab or (E) 177Lu‐DOTAGA‐PEG2‐RM26 in combination with trastuzumab. 177Lu‐DOTAGA‐PEG2‐RM26 demonstrated quantitative labeling yield at high molar activity (450 GBq/μmol), high in vivo stability (5 min pi >98% of radioligand remained when coinjected with phosphoramidon), high affinity to GRPR (KD = 0.4 ± 0.2 nM), and favorable biodistribution (1 hr pi tumor uptake was higher than in healthy tissues, including the kidneys). Therapy with 177Lu‐DOTAGA‐PEG2‐RM26 induced a significant inhibition of tumor growth. The median survival for control groups was significantly shorter than for treated groups (Group C 66 days, Group E 74 days). Trastuzumab together with radionuclide therapy significantly improved survival. No treatment‐related toxicity was observed. In conclusion, based on in vitro and in vivo characterization of the four 177Lu‐labeled PEG2‐RM26 analogs, we concluded that 177Lu‐DOTAGA‐PEG2‐RM26 was the most promising analog for TRT. Radiotherapy using 177Lu‐DOTAGA‐PEG2‐RM26 effectively inhibited tumor growth in vivo in a murine prostate cancer model. Anti‐HER2 therapy additionally improved survival. What's new? Targeted radionuclide therapy (TRT) using radiolabeled peptides seeking gastrin‐releasing peptide receptors (GRPRs) in tumors is a promising approach to treat disseminated prostate cancer. The possibility to improve the therapeutic index via combination therapies also warrants further investigation. Here, the authors developed and characterized a promising GRPR‐targeting radioligand and demonstrated its therapeutic efficacy in prostate cancer xenografts. Moreover, this study using the anti‐HER2 antibody trastuzumab presents the first in vivo proof‐of‐principle that the effects of anti‐GRPR radiotherapy can be amplified by co‐administration of anti‐HER2 treatment leading to prolonged survival.
Collapse
Affiliation(s)
- Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sara S Rinne
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Mark W Konijnenberg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", Athens, Greece
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", Athens, Greece
| | - Mohamed Altai
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mats Larhed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.,Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marion de Jong
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.,Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
89
|
Bakker IL, van Tiel ST, Haeck J, Doeswijk GN, de Blois E, Segbers M, Maina T, Nock BA, de Jong M, Dalm SU. In Vivo Stabilized SB3, an Attractive GRPR Antagonist, for Pre- and Intra-Operative Imaging for Prostate Cancer. Mol Imaging Biol 2019; 20:973-983. [PMID: 29556947 PMCID: PMC6244536 DOI: 10.1007/s11307-018-1185-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose The gastrin-releasing peptide receptor (GRPR), overexpressed on various tumor types, is an attractive target for receptor-mediated imaging and therapy. Another interesting approach would be the use of GRPR radioligands for pre-operative imaging and subsequent radio-guided surgery, with the goal to improve surgical outcome. GRPR radioligands were successfully implemented in clinical studies, especially Sarabesin 3 (SB3) is an appealing GRPR antagonist with high receptor affinity. Gallium-68 labeled SB3 has good in vivo stability, after labeling with Indium-111; however, the molecule shows poor in vivo stability, which negatively impacts tumor-targeting capacity. A novel approach to increase in vivo stability of radiopeptides is by co-administration of the neutral endopeptidase (NEP) inhibitor, phosphoramidon (PA). We studied in vivo stability and biodistribution of [111In]SB3 without/with (−/+) PA in mice. Furthermore, SPECT/MRI on a novel, state-of-the-art platform was performed. Procedures GRPR affinity of SB3 was determined on PC295 xenograft sections using [125I]Tyr4-bombesin with tracer only or with increasing concentrations of SB3. For in vivo stability, mice were injected with 200/2000 pmol [111In]SB3 −/+ 300 μg PA. Blood was collected and analyzed. Biodistribution and SPECT/MRI studies were performed at 1, 4, and 24 h postinjection (p.i.) of 2.5 MBq/200 pmol or 25 MBq/200 pmol [111In]SB3 −/+ 300 μg PA in PC-3-xenografted mice. Results SB3 showed high affinity for GRPR (IC50 3.5 nM). Co-administration of PA resulted in twice higher intact peptide in vivo vs [111In]SB3 alone. Biodistribution studies at 1, 4, and 24 h p.i. show higher tumor uptake values with PA co-administration (19.7 ± 3.5 vs 10.2 ± 1.5, 17.6 ± 5.1 vs 8.3 ± 1.1, 6.5 ± 3.3 vs 3.1 ± 1.9 % ID/g tissue (P < 0.0001)). Tumor imaging with SPECT/MRI clearly improved after co-injection of PA. Conclusions Co-administration of PA increased in vivo tumor targeting capacity of [111In]SB3, making this an attractive combination for GRPR-targeted tumor imaging.
Collapse
Affiliation(s)
- Ingrid L Bakker
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands.
| | - Sandra T van Tiel
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Joost Haeck
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Gabriela N Doeswijk
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Marcel Segbers
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Theodosia Maina
- Molecular Radiopharmacy, INSRATES, NCSR "Demokritos", Athens, Greece
| | - Berthold A Nock
- Molecular Radiopharmacy, INSRATES, NCSR "Demokritos", Athens, Greece
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Simone U Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Room No. Na2510, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| |
Collapse
|
90
|
Usmani S, Orevi M, Stefanelli A, Zaniboni A, Gofrit ON, Bnà C, Illuminati S, Lojacono G, Noventa S, Savelli G. Neuroendocrine differentiation in castration resistant prostate cancer. Nuclear medicine radiopharmaceuticals and imaging techniques: A narrative review. Crit Rev Oncol Hematol 2019; 138:29-37. [PMID: 31092382 DOI: 10.1016/j.critrevonc.2019.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Androgen Deprivation Therapy (ADT) is the primary treatment for patients suffering from relapsing or advanced prostate cancer (PC). Hormone therapy generally guarantees adequate clinical control of the disease for some years, even in those patients affected by widespread skeletal and soft tissue metastases. Despite ADT, however, most patients treated with hormones eventually progress to castration-resistant prostate cancer (CRPC), for which there are no effective treatments. This clinical reality is an open challenge to the oncologist because of those neoplasms which elaborate neuroendocrine differentiation (NED). METHODS An online search of current and past literature on NED in CRPC was performed. Relevant articles dealing with the biological and pathological basis of NED, with nuclear medicine imaging in CRPC and somatostatin treatment in NED were analyzed. EVIDENCE FROM THE LITERATURE NED may arise in prostate cancer patients in the late stages of ADT. The onset of NED offers prognostic insight because it reflects a dramatic increase in the aggressive nature of the neoplasm. Several genetic, molecular, cytological and immunohistochemical markers are associated with this transformation. Among these, overexpression of somatostatin receptors, seen through Nuclear Medicine testing, is one of the most studied. CONCLUSIONS Preliminary studies show that the overexpression of somatostatin receptors related to NED in CRPC may easily be studied in vivo with PET/CT. This finding offers a potentially useful objective for targeted therapy in CRPC. If the overexpression of SSTRs is shown to afflict a significant segment of patients with CRPC, this will open further study of possible therapeutic options based on this marker.
Collapse
Affiliation(s)
- Sharjeel Usmani
- Department of Nuclear Medicine, Kuwait Cancer Control Center Al Sabah Medical District, 70653, Kuwait
| | - Marina Orevi
- Nuclear Medicine Division, Kiryat Hadassah, POB 12000, Jerusalem 91120, Israel
| | - Antonella Stefanelli
- Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy
| | - Alberto Zaniboni
- Department of Medical Oncology, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy
| | | | - Claudio Bnà
- Radiology Division, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy
| | - Sonia Illuminati
- Radiology Division, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy
| | - Giulia Lojacono
- Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy
| | - Silvia Noventa
- Department of Medical Oncology, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy
| | - Giordano Savelli
- Nuclear Medicine Division, Fondazione Poliambulanza Istituto Ospedaliero, via L. Bissolati, 57, 25124 Brescia, Italy.
| |
Collapse
|
91
|
Lymperis E, Kaloudi A, Kanellopoulos P, de Jong M, Krenning EP, Nock BA, Maina T. Comparing Gly 11/dAla 11-Replacement vs. the in-Situ Neprilysin-Inhibition Approach on the Tumor-targeting Efficacy of the 111In-SB3/ 111In-SB4 Radiotracer Pair. Molecules 2019; 24:molecules24061015. [PMID: 30871262 PMCID: PMC6471467 DOI: 10.3390/molecules24061015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 02/08/2023] Open
Abstract
Background: The GRPR-antagonist 68Ga-SB3 visualized prostate cancer lesions in animal models and in patients. Switching radiometal from 68Ga to 111In impaired tumor targeting in mice, but coinjection of the neprilysin (NEP)-inhibitor phosphoramidon (PA) stabilized 111In-SB3 in circulation and remarkably increased tumor uptake. We herein report on the biological profile of 111In-SB4: 111In-[dAla11]SB3. Methods: The biological responses of 111In-SB3/SB4 were compared in PC-3 cells and animal models. Results: Gly11/dAla11-replacement deteriorated GRPR-affinity (SB4 IC50: 10.7 ± 0.9 nM vs. SB3 IC50: 4.6 ± 0.3 nM) and uptake in PC-3 cells (111In-SB4: 1.3 ± 0.4% vs. 111In-SB3 16.2 ± 0.8% at 1 h). 111In-SB4 was more stable than 111In-SB3, but PA-coinjection stabilized both radiotracers in peripheral mice blood. Unmodified 111In-SB3 showed higher uptake in PC-3 xenografts (8.8 ± 3.0%ID/g) vs. 111In-SB4 (3.1 ± 1.1%ID/g) at 4 h pi. PA-coinjection improved tumor uptake, with 111In-SB3 still showing superior tumor targeting (38.3 ± 7.9%ID/g vs. 7.4 ± 0.3%ID/g for 111In-SB4). Conclusions: Replacement of Gly11 by dAla11 improved in vivo stability, however, at the cost of GRPR-affinity and cell uptake, eventually translating into inferior tumor uptake of 111In-SB4 vs. unmodified 111In-SB3. On the other hand, in-situ NEP-inhibition turned out to be a more efficient and direct strategy to optimize the in vivo profile of 111In-SB3, and potentially other peptide radiotracers.
Collapse
Affiliation(s)
- Emmanouil Lymperis
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | - Aikaterini Kaloudi
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | | | - Marion de Jong
- Department of Radiology, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| | - Eric P Krenning
- Cytrotron Rotterdam BV, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| |
Collapse
|
92
|
Abstract
Prostate cancer (PCa) is the most common cancer in men worldwide, but it exhibits a highly variable biological behavior ranging from indolent to highly aggressive disease. The standard conventional imaging for staging PCa consists of CT, MRI, and bone scans, but this imaging has suboptimal accuracy for extraprostatic tumor detection, particularly in the scenario of early biochemical relapse when the prostate-specific antigen levels are still low indicating a low volume of recurrent disease. This gap between known disease (as indicated by a rising prostate-specific antigen) and the failure to detect it on conventional imaging, has led to the development of novel imaging probes most of which have positron emitting radioactive tags. In the last decade, multiple PET probes have demonstrated promising performance in detecting sites of recurrence and extent of disease in patients with PCa. The landscape of available PET radiotracers is changing rapidly and includes radiolabeled choline, anti1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (18F-fluciclovine), bombesin, dihydrotestosterone, and prostate-specific membrane antigen (PSMA) ligands, among others. Of these, radiolabeled PSMA-PET agents have shown the most encouraging results in terms of sensitivity and are likely to become universally available for imaging PCa within a few years Other PET radiotracers such as bombesin-based radiotracers and antagonist of gastrin releasing-peptide receptor (RM2) are emerging as possible alternatives for PCa imaging. This review article discusses the current and near-future of PET molecular imaging probes.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Program, National Cancer Institute, NIH. Bethesda, MD
| | - Liza M Lindenberg
- Molecular Imaging Program, National Cancer Institute, NIH. Bethesda, MD
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, NIH. Bethesda, MD.
| |
Collapse
|
93
|
Zhuo L, Yang X, Liao W, Wang J, Wang H, Lv M, Wang G, Song H, Feng Y, Chen Y, Wei H, Yang Y, Zhao P. Comparative cell uptake study of FITC-/177Lu-labeled RM26 monomer, dimer and trimer on PC-3: improving binding affinity of gastrin releasing peptide receptor (GRPR) antagonist via bivalency/trivalency. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-018-6396-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
94
|
Guillou A, Lima LMP, Esteban-Gómez D, Le Poul N, Bartholomä MD, Platas-Iglesias C, Delgado R, Patinec V, Tripier R. Methylthiazolyl Tacn Ligands for Copper Complexation and Their Bifunctional Chelating Agent Derivatives for Bioconjugation and Copper-64 Radiolabeling: An Example with Bombesin. Inorg Chem 2019; 58:2669-2685. [PMID: 30689368 DOI: 10.1021/acs.inorgchem.8b03280] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We present here the synthesis of two new bifunctionalized azachelators, no2th-EtBzNCS and Hno2th1tha, as bioconjugable analogues of two previously described di- and trimethylthiazolyl 1,4,7-triazacyclononane (tacn) ligands, no2th and no3th, for potential uses in copper-64 (64Cu) positron emission tomography imaging. The first one bears an isothiocyanate group on the remaining free nitrogen atom of the tacn framework, while the second one presents an additional carboxylic function on one of the three heterocyclic pendants. Their syntheses required regiospecific N-functionalization of the macrocycles. In order to investigate their suitability for in vivo applications, a complete study of their copper(II) chelation was performed. The acid-base properties of the ligands and their thermodynamic stability constants with copper(II) and zinc(II) cations were determined using potentiometric techniques. Structural studies were conducted in both solution and the solid state, consolidated by theoretical calculations. The kinetic inertness in an acidic medium of both copper(II) complexes was determined by spectrophotometry, while cyclic voltammetry experiments were performed to evaluate the stability at the copper(I) redox state. UV-vis, NMR (of the zinc complexes), electron paramagnetic resonance spectroscopy, and density functional theory studies showed excellent agreement between the solution structures of the complexes and their crystallographic data. These investigations unambiguously prove that these bifunctional derivatives display similar coordination properties as their no2th and no3th counterparts, opening the door to targeted bioapplications. The no2th-EtBzNCS and Hno2th1tha ligands were then conjugated to a bombesin antagonist peptide for targeting the gastrin-releasing peptide receptor (GRPr). To highlight the potential of the two chelators for radiopharmaceutical development, the 64Cu-radiolabeling properties, in vitro stability, and binding affinity to GRPr of the corresponding bioconjugates were determined. Altogether, the results of this work warrant the further development of 64Cu-based radiopharmaceuticals comprising our novel bifunctional chelators.
Collapse
Affiliation(s)
- Amaury Guillou
- UFR des Sciences et Techniques , UMR-CNRS 6521, Université de Bretagne Occidentale , 6 avenue Victor le Gorgeu, C.S. 93837 , 29238 Brest Cedex 3 , France
| | - Luís M P Lima
- Instituto de Tecnologia Química e Biológica António Xavier , Universidade Nova de Lisboa , Avenida da República , 2780-157 Oeiras , Portugal
| | - David Esteban-Gómez
- Departamento de Química, Facultade de Ciencias & Centro de Investigaciones Científicas Avanzadas , Universidade da Coruña , 15071 A Coruña , Spain
| | - Nicolas Le Poul
- UFR des Sciences et Techniques , UMR-CNRS 6521, Université de Bretagne Occidentale , 6 avenue Victor le Gorgeu, C.S. 93837 , 29238 Brest Cedex 3 , France
| | - Mark D Bartholomä
- Department of Nuclear Medicine , Saarland University-Medical Center , Kirrbergerstrasse , 66421 Homburg , Germany
| | - Carlos Platas-Iglesias
- Departamento de Química, Facultade de Ciencias & Centro de Investigaciones Científicas Avanzadas , Universidade da Coruña , 15071 A Coruña , Spain
| | - Rita Delgado
- Instituto de Tecnologia Química e Biológica António Xavier , Universidade Nova de Lisboa , Avenida da República , 2780-157 Oeiras , Portugal
| | - Véronique Patinec
- UFR des Sciences et Techniques , UMR-CNRS 6521, Université de Bretagne Occidentale , 6 avenue Victor le Gorgeu, C.S. 93837 , 29238 Brest Cedex 3 , France
| | - Raphaël Tripier
- UFR des Sciences et Techniques , UMR-CNRS 6521, Université de Bretagne Occidentale , 6 avenue Victor le Gorgeu, C.S. 93837 , 29238 Brest Cedex 3 , France
| |
Collapse
|
95
|
Cellular internalization of a cell-penetrating peptide conjugated gastrin-releasing peptide receptor antagonist. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-019-06418-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
96
|
Akbar MJ, Lukasewicz Ferreira PC, Giorgetti M, Stokes L, Morris CJ. Bombesin receptor-targeted liposomes for enhanced delivery to lung cancer cells. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2019; 10:2553-2562. [PMID: 31921534 PMCID: PMC6941431 DOI: 10.3762/bjnano.10.246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/03/2019] [Indexed: 05/09/2023]
Abstract
Background: Gastrin-releasing peptide is a member of the bombesin family of peptides. Its cognate receptor, gastrin releasing peptide receptor (GRPR), is widely expressed in cancers of the lung, pancreas and ovaries. Gastrin releasing peptide (GRP) is an autocrine growth factor in small cell lung cancer, which has very poor patient outcomes. High affinity antagonist peptides have been developed for in vivo cancer imaging. In this report we decorated pegylated liposomes with a GRPR antagonist peptide and studied its interaction with, and accumulation within, lung cancer cells. Results: An N-terminally cysteine modified GRPR antagonist (termed cystabn) was synthesised and shown to inhibit cell growth in vitro. Cystabn was used to prepare a targeted 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPE-PEG2000) lipid conjugate that was formulated into liposomes. The liposomes displayed desirable colloidal properties and good stability under storage conditions. Flow cytometric and microscopic studies showed that fluorescently labelled cystabn-decorated liposomes accumulated more extensively in GRPR over-expressing cells than matched liposomes that contained no cystabn targeting motif. Conclusion: The use of GRPR antagonistic peptides for nanoparticle targeting has potential for enhancing drug accumulation in resistant cancer cells.
Collapse
Affiliation(s)
| | | | | | - Leanne Stokes
- School of Pharmacy, University of East Anglia, Norwich, UK
| | | |
Collapse
|
97
|
Lv M, Zhao P, Zhuo L, Liao W, Wang H, Yang X, Wang J, Wang G, Song H, Feng Y, Chen Y, Yang Y, Wei H. Binding and cytotoxicity of 131I-labeled gastrin-releasing peptide receptor antagonists modified by cell penetrating peptides. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-018-6307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
98
|
Gnesin S, Cicone F, Mitsakis P, Van der Gucht A, Baechler S, Miralbell R, Garibotto V, Zilli T, Prior JO. First in-human radiation dosimetry of the gastrin-releasing peptide (GRP) receptor antagonist 68Ga-NODAGA-MJ9. EJNMMI Res 2018; 8:108. [PMID: 30543050 PMCID: PMC6291411 DOI: 10.1186/s13550-018-0462-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/26/2018] [Indexed: 12/15/2022] Open
Abstract
Background Gastrin-releasing peptide receptor antagonists have promise in theranostics of several highly incident tumours, including prostate and breast. This study presents the first human dosimetry of 68Ga-NODAGA-MJ9 in the first five consecutive patients with recurrent prostate cancer included in a dual-tracer positron emission tomography (PET) protocol. Five male patients with biochemical relapse of prostate adenocarcinoma underwent four whole-body time-of-flight PET/CT scans within 2 h after tracer injection. To be used as input in OLINDA/EXM 2.0, time-integrated activity coefficients were derived from manually drawn regions of interest over the following body regions: brain, thyroid, lungs, heart, liver, gallbladder, spleen, stomach, kidneys, adrenals, red marrow, pancreas, intestines, urinary bladder and whole body. Organ absorbed doses and effective dose (ED) were calculated with OLINDA/EXM 2.0 using the NURBS voxelized phantoms adjusted to the ICRP-89 organ masses and ICRP103 tissue-weighting factors. Additional absorbed dose estimations were performed with OLINDA/EXM 1.1 to be comparable with similar previous publications. Results The body regions receiving the highest absorbed doses were the pancreas, the urinary bladder wall, the small intestine and the kidneys (260, 69.8, 38.8 and 34.8 μGy/MBq respectively). The ED considering a 30-min urinary voiding cycle was 17.6 μSv/MBq in male patients. The increment of voiding time interval produced a significant increase of absorbed doses in bladder, prostate and testes, as well as an increase of ED. ED also increased if calculated with OLINDA/EXM 1.1. These results have been discussed in view of similar publications on bombesin analogues or on other commonly used theranostic peptides. Conclusions The pancreas is the most irradiated organ after the injection of 68Ga-NODAGA-MJ9, followed by the urinary bladder wall, the small intestine and the kidneys. ED is in the same range of other common 68Ga-labelled peptides. Differences with similarly published studies on bombesin analogues exist, and are mainly dependent on the methodology used for absorbed dose calculations. Trial registration Clinicaltrial.Gov identifier: NCT02111954, posted on 11/042014. Electronic supplementary material The online version of this article (10.1186/s13550-018-0462-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital, Rue du Grand-Pré 1, 1007, Lausanne, Switzerland.
| | - Francesco Cicone
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Periklis Mitsakis
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Axel Van der Gucht
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Sébastien Baechler
- Institute of Radiation Physics, Lausanne University Hospital, Rue du Grand-Pré 1, 1007, Lausanne, Switzerland
| | - Raymond Miralbell
- Department of Radiation Oncology, University Hospital of Geneva and Geneva University, Geneva, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, University Hospital of Geneva and Geneva University, Geneva, Switzerland
| | - Thomas Zilli
- Department of Radiation Oncology, University Hospital of Geneva and Geneva University, Geneva, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
99
|
Zang J, Mao F, Wang H, Zhang J, Liu Q, Peng L, Li F, Lang L, Chen X, Zhu Z. 68Ga-NOTA-RM26 PET/CT in the Evaluation of Breast Cancer: A Pilot Prospective Study. Clin Nucl Med 2018; 43:663-669. [PMID: 30036253 PMCID: PMC6076351 DOI: 10.1097/rlu.0000000000002209] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND This prospective pilot study investigated the value of Ga-NOTA-RM26, an antagonist targeting gastrin-releasing peptide receptor, in evaluation of breast cancer. METHODS Thirty-five women in suspicion of breast cancer based on mammography or ultrasonography were recruited with informed consent. They underwent PET/CT scans 30 minutes after intravenous injection of Ga-NOTA-RM26 in a dose of 1.85 MBq (0.05 mCi) per kilogram body weight within 1 week before surgery. The Ga-NOTA-RM26 uptake was correlated with the pathological and immunohistochemical findings. RESULTS Ga-NOTA-RM26 positivity was found correlated with estrogen receptor (ER) expression (P = 0.006) and menstrual status (P = 0.019). In 34 patients diagnosed with breast cancer, the SUVmax was found significantly higher in the ER-positive breast cancer (4.97 ± 1.89) than in the ER-negative breast cancer (2.78 ± 0.65, P < 0.001). Ga-NOTA-RM26 was also found accumulated in normal breast tissue, with the SUVmax significantly higher in patients at the secretory phase of menstrual cycle (2.27 ± 0.71) than in those at the nonsecretory phase (1.59 ± 0.49, P = 0.017) and postmenopause (1.43 ± 0.44, P = 0.002). If the secretory phase patients were excluded, the sensitivity, specificity, and accuracy for differentiation of breast cancer from breast tissue increased from 85.3%, 86.8%, and 86.1% to 100%, 90.9%, and 95.5%, respectively. CONCLUSIONS This pilot study indicates that the diagnostic accuracy of Ga-NOTA-RM26 PET/CT in breast cancer may correlate with ER expression and menstrual status of the patient. It may be better to avoid performing this examination during the menstrual secretory phase to reduce physiological uptake in normal breast tissue.
Collapse
Affiliation(s)
- Jie Zang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Feng Mao
- Department of Breast Surgery, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
| | - Hao Wang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Jingjing Zhang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Qingxing Liu
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Li Peng
- Department of Breast Surgery, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Lixin Lang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| |
Collapse
|
100
|
Xu H, Bandari RP, Lee L, Li R, Yu P, Smith CJ, Ma L. Design, Synthesis, and in Vitro and in Vivo Evaluation of High Affinity and Specificity Near-Infrared Fluorescent Bombesin Antagonists for Tumor Imaging. J Med Chem 2018; 61:7657-7670. [DOI: 10.1021/acs.jmedchem.8b00614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hang Xu
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Department of Chemical Engineering, University of Missouri, Columbia, Missouri 65211, United States
| | - Rajendra P. Bandari
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Li Lee
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri 65211, United States
| | - Ran Li
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Department of Stomatology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ping Yu
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri 65211, United States
| | - Charles J. Smith
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Lixin Ma
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|