1
|
Meng L, Fang J, Lin X, Zhuang R, Huang L, Li Y, Zhang X, Guo Z. Development of radioligands with an albumin-binding moiety of 4-(P-Iodophenyl) butyric acid for theranostic applications. J Control Release 2025; 382:113757. [PMID: 40262707 DOI: 10.1016/j.jconrel.2025.113757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
The rapid clearance of imaging probes from blood circulation is beneficial for receptor imaging, as it minimizes non-target tissue exposure and improves tumor-to-background contrast. However, this rapid clearance can hinder radioligand therapy by limiting tumor uptake of radiolabeled compounds. An optimal blood half-life is crucial, as it enhances the uptake of radiolabeled compounds in targets, improving tumor uptake and retention of small molecule drugs, and thus therapeutic outcomes. To address this, strategies to extend blood half-life have been developed, with the addition of an albumin-binding moiety (ABM) being particularly effective. Among these, 4-(p-iodophenyl)butyric acid (IPBA) has emerged as a versatile ABM for radiopharmaceutical design. IPBA conjugation has successfully enhanced tissue distribution profiles across various cancer types. This review highlights recent progress in the design, radiosynthesis, and application of IPBA-based small molecular radioligands, providing insights for future clinical development of IPBA-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Lingxin Meng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jianyang Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xiaoru Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Rongqiang Zhuang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Lumei Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Xianzhong Zhang
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhide Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China; Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.
| |
Collapse
|
2
|
Hasegawa T, Nakashima K, Tarumizu Y, Tada M, Maya Y, Watanabe H, Ono M. Development of Novel Radiotheranostic Ligand with Positively Charged Unit Targeting Prostate-Specific Membrane Antigen. J Med Chem 2025; 68:10190-10202. [PMID: 40358362 DOI: 10.1021/acs.jmedchem.5c00370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Prostate-specific membrane antigen (PSMA) is an ideal target of prostate cancer (PCa) for theranostics, combining diagnosis and therapy in the field of nuclear medicine. [177Lu]Lu-PSMA-617 is a gold standard in PSMA-targeting radioligands, whereas its rapid clearance from the tumor and high uptake in the kidney may compromise the efficacy of theranostics. In this study, we developed novel PSMA-targeting radioligands, [111In]In/[225Ac]Ac-PDI2 and [111In]In/[225Ac]Ac-PDI4, by introducing a positively charged diethylenetriamine (PEI2) or tetraethylenepentamine (PEI4) structure, respectively, to PSMA-617. In the biodistribution study, higher tumor retention and lower renal uptake of [111In]In-PDI2 and [111In]In-PDI4 were observed than those of [111In]In-PSMA-617, and [111In]In-PDI2 exhibited higher tumor-residualizing properties than [111In]In-PDI4. [111In]In-PDI2 and [111In]In-PDI4 clearly visualized PSMA-expressing tumors by single photon emission computed tomography/computed tomography (SPECT/CT). The administration of [225Ac]Ac-PDI2 led to a higher antitumor effect than [225Ac]Ac-PDI4 and [225Ac]Ac-PSMA-617. These findings suggest the utility of [111In]In/[225Ac]Ac-PDI2 as theranostic ligands for PCa.
Collapse
Affiliation(s)
- Takuma Hasegawa
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuta Tarumizu
- Research Center, Nihon Medi-Physics Co., Ltd., Chiba 299-0266, Japan
| | - Masatoshi Tada
- Research Center, Nihon Medi-Physics Co., Ltd., Chiba 299-0266, Japan
| | - Yoshifumi Maya
- Research Center, Nihon Medi-Physics Co., Ltd., Chiba 299-0266, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
3
|
Huynh TT, Feng Y, Banks RL, Vaidyanathan G, Zalutsky MR. Enhancing the therapeutic index of [ 211At]YF2 with iodo pseudo carrier: A simple strategy for reducing accumulation in kidneys, salivary and lacrimal glands. Nucl Med Biol 2025; 146-147:109028. [PMID: 40393125 DOI: 10.1016/j.nucmedbio.2025.109028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/18/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Low-molecular-weight (LMW) PSMA-targeted agents have enjoyed success; however, their off-target toxicity in normal tissues such as kidneys, salivary gland and lacrimal gland can be dose limiting. Herein, we have evaluated the effect of co-administration of the non-radioactive iodo pseudo carrier, iodo YF2, on the normal tissue uptake of [211At]YF2 in xenografted mice. The potential implications for clinical translation of these studies were investigated by evaluating the binding of LMW PSMA-targeted agents to murine and human PSMA. METHODS [211At]YF2 was synthesized following established protocols. Groups of 5 mice bearing subcutaneous PSMA+ PC3 PIP xenografts received [211At]YF2 co-injected with varying i.v. doses of iodo YF2 (0-2 nmol), and the biodistribution was evaluated at 1 h post injection (p.i.). In another study, the biodistribution of [211At]YF2 alone and [211At]YF2 co-administered with 1.5 nmol iodo YF2 was evaluated at 1 and 8 h p.i. Bead-based radioligand binding assays were conducted for [131I]YF2 and several other LMW agents to compare their binding to human and murine PSMA. RESULTS At 1 h, no significant difference was seen in kidney uptake of [211At]YF2 at iodo YF2 concentrations of 0, 0.05 and 0.1 nmol (p > 0.05), but renal accumulations significantly reduced by co-administering 2.0 nmol iodo YF2 (p < 0.0001). Decreases in salivary and lacrimal gland uptake also were observed at 1 h for [211At]YF2 co-injected with 0.1 nmol iodo YF2 (p < 0.05). A follow-up study revealed a kidney uptake of 1.8 ± 0.4 % ID/g for [211At]YF2 with 1.5 nmol iodo YF2, compared to 46.5 ± 7.7 % ID/g for [211At]YF2 alone at 8 h. Tumor uptake showed no significant difference (p > 0.05) between [211At]YF2 alone (17.1 ± 8.8 % ID/g at 1 h; 13.6 ± 5.3 % ID/g at 8 h) and [211At]YF2 plus 1.5 nmol iodo YF2 (12.8 ± 2.7 % ID/g at 1 h; 14.1 ± 3.2 % ID/g at 8 h). Bead-based radioligand binding assays showed that [131I]YF2 has the highest binding fractions to both human PSMA (94.1 ± 0.1 %) and murine PSMA (92.5 ± 0.2 %) with minimal differences between the two, while [177Lu]PSMA-617 had the greatest species-dependent disparity with a binding fraction of 90.5 ± 0.3 % to human PSMA and 60.9 ± 1.4 % to murine PSMA. CONCLUSIONS Co-administration of iodo YF2 reduced kidney uptake of [211At]YF2 and decreased accumulation in normal tissues with no significant change in tumor uptake. In some cases, there was a significant difference in binding to human and murine PSMA among LMW PSMA-targeted agents suggesting that particularly for some agents, applying mouse data to predict human dosimetry must be done with caution.
Collapse
Affiliation(s)
- Truc T Huynh
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca L Banks
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
4
|
Beyer D, Vaccarin C, Schmid JV, Deberle LM, Deupi X, Schibli R, Müller C. Design and Preclinical Evaluation of Novel uPAR-Targeting Radiopeptides Modified with an Albumin-Binding Entity. Mol Pharm 2025. [PMID: 40326657 DOI: 10.1021/acs.molpharmaceut.5c00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Several studies have focused on the development and application of radiolabeled DOTA-AE105 for targeting the urokinase-type plasminogen activator receptor (uPAR), which is expressed on various cancer types. The aim of this project was to design and evaluate novel uPAR-targeting radiopeptides with improved pharmacokinetic properties in view of their therapeutic application. Five peptides (uPAR-01, uPAR-02, uPAR-03, uPAR-04, and uPAR-05) were synthesized based on the AE105 peptide backbone, a DOTA chelator, and the 4-(p-iodophenyl)butanoate moiety as an albumin binder. The peptides were obtained in 20-29 synthetic steps using solid-phase peptide synthesis with a 6-34% overall yield. In saline, the 177Lu-labeled peptides (100 MBq/nmol) were stable (>93% intact radiopeptides) in the presence of l-ascorbic acid over 24 h. The new radiopeptides were also stable (>98% intact radiopeptides) in mouse and human blood plasma, while only ∼13% of [177Lu]Lu-DOTA-AE105 was intact after a 4 h incubation period. The uPAR-binding affinities (KD values) determined with uPAR-transfected human embryonic kidney cells (HEK-uPAR) ranged from 10 to 57 nM and were, thus, similar to that of [177Lu]Lu-DOTA-AE105 (KD: 20 ± 1 nM). Compared to [177Lu]Lu-DOTA-AE105, the radiopeptides showed the anticipated increased binding affinity to plasma proteins both in mouse (31- to 104-fold) and human blood plasma (43- to 136-fold). The tissue distribution of the novel radiopeptides in nude mice bearing HEK-uPAR xenografts showed substantial activity retention in the blood (12-16% IA/g and 4.5-13% IA/g at 4 and 24 h p.i., respectively), while [177Lu]Lu-DOTA-AE105 was rapidly cleared (<0.1% IA/g at 4 h p.i.). As a result, the accumulation of the new radiopeptides in HEK-uPAR xenografts (3.6-11% and 3.1-10% IA/g at 4 and 24 h p.i., respectively) was increased in comparison to that of [177Lu]Lu-DOTA-AE105 (<1% IA/g at 4 h p.i.). Importantly, the metabolic stability of the new radiopeptides in mice was enhanced as compared to that of [177Lu]Lu-DOTA-AE105. [177Lu]Lu-uPAR-02 showed the most promising tissue distribution profile with over 10-fold higher activity retention in the HEK-uPAR xenograft than observed after injection of [177Lu]Lu-DOTA-AE105. As a result, the xenograft-to-kidney ratio of [177Lu]Lu-uPAR-02 was >3-fold higher than that of [177Lu]Lu-DOTA-AE105.
Collapse
Affiliation(s)
- Darja Beyer
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Christian Vaccarin
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Jerome V Schmid
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Luisa M Deberle
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Xavier Deupi
- Condensed Matter Theory Group, PSI Center for Scientific Computing, Theory, and Data, 5232 Villigen-PSI, Switzerland
- Laboratory of Biomolecular Research, PSI Center for Life Sciences, 5232 Villigen-PSI, Switzerland
- Swiss Institute of Bioinformatics (SIB), 1015 Lausanne, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
5
|
Yang H, Wang J, Wen X, Guo H, Jakobsson V, Zhao T, Zeng F, Shen H, Zhang H, Liu X, Qin Y, Li X, Xiong H, Zhou Z, Zhang J, Chen X. Dansylated Amino Acid-Modified Long-Acting PSMA Derivatives 68Ga/ 177Lu-LNC1011 as Prostate Cancer Theranostics. J Nucl Med 2025; 66:739-747. [PMID: 40113222 DOI: 10.2967/jnumed.124.268959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radiopharmaceutical therapy has demonstrated promising potential for treating metastatic castration-resistant prostate cancer. Recently, albumin-binding motif-modified PSMA radioligands with prolonged blood circulation were developed to improve tumor uptake and therapeutic effectiveness, properties which, however, were associated with an increased risk of bone marrow toxicity. This study presents new PSMA-targeted radioligands incorporating dansylated amino acids as relatively weak and preferable albumin binders to achieve a fine balance between increased tumor accumulation, safety, and diagnostic efficacy, facilitating a unified approach to theranostics within a single molecular framework. Methods: Three novel PSMA ligands ([68Ga]Ga-Dan-Gly-PSMA, [68Ga]Ga-Dan-Nva-PSMA, and [68Ga]Ga-Dan-Phe-PSMA, denoted as [68Ga]Ga-LNC1011) were synthesized with dansylated amino acids and measured the albumin-binding properties with human serum albumin through ultrafiltration experiments. Binding affinity and PSMA-targeting specificity were investigated using a saturation binding assay and cell uptake in the PSMA-induced prostate cancer 3 cell line (PC3-PIP). PET imaging in PC3-PIP tumor-bearing mice was performed to evaluate the preclinical pharmacokinetics and diagnostic efficiency of 68Ga-labeled PSMA ligands. Tumor uptake of [177Lu]Lu-LNC1011 was evaluated through SPECT/CT imaging and biodistribution studies. Radiopharmaceutical therapy studies were conducted to systematically assess the therapeutic effect of the radioligand. Results: Three novel PSMA radioligands ([68Ga]Ga-Dan-Gly-PSMA, [68Ga]Ga-Dan-Nva-PSMA, and [68Ga]Ga-LNC1011) with various dansylated amino acids were successfully synthesized with a radiochemical yield greater than 97%. In the PC3-PIP xenograft tumor model, the tumor/heart, tumor/liver, tumor/kidney, and tumor/muscle ratios were 9.82 ± 2.35, 12.42 ± 3.71, 4.36 ± 0.29, and 52.88 ± 12.08 at 4 h after injection, respectively. Biodistribution studies confirmed the significantly higher tumor uptake of [177Lu]Lu-LNC1011 (127.36 ± 16.95 %ID/g) over [177Lu]Lu-PSMA-617 (17.44 ± 6.29 %ID/g) at 4 h after injection, and no decrease was measured for the [177Lu]Lu-LNC1011 at up to 72 h after injection, which was corroborated with SPECT imaging. A single injection of 9.3 MBq of [177Lu]Lu-LNC1011 achieved 89.43% inhibition of tumor growth, equivalent to 18.5 MBq of [177Lu]Lu-PSMA-617 (90.87%). [68Ga]Ga-LNC1011 PET/CT scans of patients with metastatic castration-resistant prostate cancer identified as many lesions as [68Ga]Ga-PSMA-11 did, confirming its diagnostic efficacy. Conclusion: 68Ga/177Lu-LNC1011, characterized by high tumor uptake and retention along with timely clearance from normal organs and tissues, thus emerges as a promising single-molecule theranostic radioligand.
Collapse
Affiliation(s)
- Hongzhang Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jiarou Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xuejun Wen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Huifeng Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Fantian Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Huaxiang Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Heng Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Xiaomin Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Yatong Qin
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Xinyi Li
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Hehe Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Zijian Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang'an Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, China
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore;
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore;
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore; and
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Proteos, Singapore
| |
Collapse
|
6
|
Sobral MC, Mota SI, Oliveira PJ, Urbano AM, Paulo A. Two Targets, One Mission: Heterobivalent Metal-Based Radiopharmaceuticals for Prostate Cancer Imaging and Therapy. ChemMedChem 2025:e2500128. [PMID: 40117450 DOI: 10.1002/cmdc.202500128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 03/23/2025]
Abstract
Prostate cancer (PCa) is a significant healthcare challenge, associated with considerable mortality and morbidity among men, particularly in developed countries. PCa mortality and morbidity are primarily related to its most advanced form, metastatic castration-resistant PCa (mCRPC), for which there is presently no cure. Therefore, novel therapeutic approaches to increase mCRPC survival are critically needed. Due to PCa tumor heterogeneity and a complex tumor microenvironment, the efficacy of single-target radiopharmaceuticals, such as the Food and Drug Administration-approved [177Lu]Lu-PSMA-617, is currently under reassessment. The design and development of PCa dual-target radiopharmaceuticals have garnered considerable attention, due to their benefits over single-target counterparts, namely increased therapeutic specificity and efficacy, as well as the ability to overcome the challenge of inconsistent tumor visualization caused by variable receptor expression across diverse lesions, thereby enabling more comprehensive imaging. Several PCa biomarkers are currently being investigated as potential targets for dual-target radiopharmaceuticals, including prostate-specific membrane antigen, gastrin-releasing peptide receptor, integrin αvβ3 receptor, fibroblast activation protein, sigma-1 receptor, as well as albumin, the radiosensitive cell nucleus, and mitochondria. This review explores recent advancements in heterobivalent metal-based radiopharmaceuticals for dual targeting in PCa, highlighting their significance in theranostic and personalized medicine.
Collapse
Affiliation(s)
- Margarida C Sobral
- Institute of Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197, Cantanhede, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548, Coimbra, Portugal
- Molecular Physical Chemistry R&D Unit, Department of Life Sciences, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Sandra I Mota
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197, Cantanhede, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197, Cantanhede, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Ana M Urbano
- Molecular Physical Chemistry R&D Unit, Department of Life Sciences, University of Coimbra, 3000-456, Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, 3001-301, Coimbra, Portugal
| | - António Paulo
- C2TN -Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, University of Lisboa, 2695-066, Lisboa, Portugal
- Department of Engineering and Nuclear Sciences, Instituto Superior Técnico, University of Lisboa, 2695-066, Lisboa, Portugal
| |
Collapse
|
7
|
Rosales-Barrios C, González-Sánchez ZI, Zuliani A, Jiménez-Vacas JM, Luque RM, Pozo D, Khiar N. PSMA-targeted delivery of docetaxel in prostate cancer using small-sized PDA-based micellar nanovectors. J Control Release 2025; 379:890-905. [PMID: 39864631 DOI: 10.1016/j.jconrel.2025.01.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
In this study, we present the first comparative analysis of active and passive drug delivery systems for docetaxel (DTX) in prostate cancer using supramolecular self-assembled micellar nanovectors. Specifically, we developed two novel micelles based on polydiacetylenic amphiphiles (PDA) for passive and active targeting. The active targeting micelles were designed with a prostate-specific membrane antigen (PSMA) ligand, ACUPA, to facilitate recognition by PSMA-positive cancer cells. These PDA-based micelles feature a well-defined structure with a hydrophobic PDA core and a surface functionalized with PEG, and for active targeting, ACUPA. Our micelles demonstrated excellent encapsulation capacity, significantly improving DTX solubility in water, a crucial factor for clinical drug use. In vitro studies confirmed the safety and cytotoxic profiles of both systems, with ACUPA-functionalized micelles showing notable internalization into PSMA-positive LNCaP cells, mediated through the PSMA-ACUPA interaction. In vivo imaging revealed preferential accumulation of ACUPA-functionalized nanomicelles in LNCaP xenograft tumors, suggesting enhanced retention via specific ACUPA-PSMA interactions and active uptake by LNCaP cells. Notably, Balb/c-Foxn1nu/nu early in vivo studies showed a marked reduction in tumor volume and tumor expression levels of proliferation, cell cycle progression, cell survival and anti-apoptotic markers with DTX-loaded micelles functionalized with ACUPA compared to those without ACUPA. Overall, our studies collect initial evidence regarding the feasibility of supramolecular self-assembly of ACUPA-PDA-based nanomicelles for PSMA-targeted drug chemotherapy delivery developments.
Collapse
Affiliation(s)
- Cristian Rosales-Barrios
- Asymmetric Synthesis and Functional Nanosystems Group (Art&Fun), Institute of Chemical Research (IIQ), CSIC-University of Seville, C/ Américo Vespucio 49, 41092 Seville, Spain
| | - Zaira I González-Sánchez
- Department of Integrative Pathophysiology and Therapies, Andalusian Centre for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-Universidad Pablo de Olavide-Universidad de Sevilla, Av. Americo Vespucio 24, Seville 41092, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology University of Seville, Av. Sánchez Pizjuan s/n, 41009 Seville, Spain; Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra (PUCMM), Hwy. Duarte km 1.5, Santiago de los Caballeros, 822, Dominican Republic
| | - Alessio Zuliani
- Asymmetric Synthesis and Functional Nanosystems Group (Art&Fun), Institute of Chemical Research (IIQ), CSIC-University of Seville, C/ Américo Vespucio 49, 41092 Seville, Spain
| | - Juan M Jiménez-Vacas
- Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University Hospital Reina Sofía (HURS), Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av. Menéndez Pidal s/n, Córdoba 14004, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Campus Rabanales, 14004 Córdoba, Spain
| | - Raul M Luque
- Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University Hospital Reina Sofía (HURS), Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av. Menéndez Pidal s/n, Córdoba 14004, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Campus Rabanales, 14004 Córdoba, Spain
| | - David Pozo
- Department of Integrative Pathophysiology and Therapies, Andalusian Centre for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-Universidad Pablo de Olavide-Universidad de Sevilla, Av. Americo Vespucio 24, Seville 41092, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology University of Seville, Av. Sánchez Pizjuan s/n, 41009 Seville, Spain
| | - Noureddine Khiar
- Asymmetric Synthesis and Functional Nanosystems Group (Art&Fun), Institute of Chemical Research (IIQ), CSIC-University of Seville, C/ Américo Vespucio 49, 41092 Seville, Spain.
| |
Collapse
|
8
|
Li P, Yang Z, Li Y, Yu J, Wang Z, Nie J, Liu X, Hou W, Zhao Y, Dai D, Li Y. Synthesis and Imaging of Novel CDK19-Targeted Tracers Incorporating an Albumin-Binding Moiety. J Labelled Comp Radiopharm 2025; 68:e4130. [PMID: 40077993 DOI: 10.1002/jlcr.4130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Cyclin-dependent kinase 19 (CDK19) is a potential target for the diagnosis and treatment of prostate cancer. We have previously studied a series of CDK19-targeted PET tracers, but in-depth drug optimization is needed to improve the physiochemical properties of such large and polar tracers. The albumin strategy has received widespread attention in recent years, and we synthesized 68Ga-IRM-14a and 68Ga-IRM-14b based on the strategy. After in vivo imaging studies in mice, we found that introducing albumin moiety will significantly change the physicochemical properties of existing large polarity tracers, thereby increasing tissue uptake and retention, which is beneficial for future treatment. In short, the albumin strategy will be an important strategy in the field of radiopharmaceutical optimization.
Collapse
Affiliation(s)
- Panfeng Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhao Yang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Yanli Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiang Yu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ziyang Wang
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Jiaci Nie
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaoman Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenbin Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
9
|
Zhou H, Zhong J, Liu Y, Peng S, Yan Q, Wang L, Zhong Y, Hu K. Development of ibuprofen-modified fibroblast activation protein radioligands to improve cancer therapy. Eur J Med Chem 2025; 283:117115. [PMID: 39626520 DOI: 10.1016/j.ejmech.2024.117115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025]
Abstract
FAP-targeting radioligands are used in cancer diagnosis and therapy, but their effectiveness is limited by poor tumor uptake and retention. This study aimed to develop new radioligands using an optimized amino acid linker and ibuprofen for better pharmacokinetics. Three novel quinoline-based FAP ligands with an ibuprofen moiety were synthesized and radiolabeled with gallium-68 and lutetium-177. The synthesized FAP ligands FAPI-Ibu1, 2, 3 showed high binding affinity for FAP, with IC50 values of 1.17 ± 0.09, 0.29 ± 0.06, and 0.78 ± 0.12 nM, respectively. 177Lu-labeled FAP ligands showed stability in vitro and demonstrated significant binding to human plasma proteins as well as FAP specificity. PET imaging and biodistribution studies of 68Ga- or 177Lu-labeled FAPI-Ibu1, 2, 3 revealed improved tumor accumulation and retention. Dosimetry calculation showed that [177Lu]Lu-FAPI-Ibu3 delivered a 9.9-fold higher absorbed dose to tumor than [177Lu]Lu-FAPI-04, but only 2.6-fold higher absorbed dose to kidneys leading to 3.8-fold improvement in the tumor-to-kidney absorbed dose ratios. In the endoradiotherapy study, 18.5 MBq of [177Lu]Lu-FAPI-Ibu3 resulted in longer median survival than the equivalent dose of [177Lu]Lu-FAPI-04 (22 vs 16 days). Three ibuprofen-modified FAP radioligands significantly improved tumor uptake, retention, and growth suppression compared to [177Lu]Lu-FAPI-04, with [177Lu]Lu-FAPI-Ibu3 emerging as the most promising candidate for further clinical translational studies.
Collapse
Affiliation(s)
- Hui Zhou
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Jiawei Zhong
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Nuclear Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Liu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Simin Peng
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qingsong Yan
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lijuan Wang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuhua Zhong
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Kongzhen Hu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
10
|
Ritt P, Fernández R, Soza-Ried C, Nicolai H, Amaral H, Krieger K, Mapanao AK, Rotger A, Zhernosekov K, Schibli R, Müller C, Kramer V. Biodistribution and dosimetry of [ 177Lu]Lu-SibuDAB in patients with metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07102-8. [PMID: 39894880 DOI: 10.1007/s00259-025-07102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Several prostate-specific membrane antigen (PSMA) radiopharmaceuticals have been used for the treatment of metastatic, castration-resistant prostate cancer (mCRPC). In an attempt to improve the tumour accumulation, new PSMA ligands were developed with an albumin-binding entity to enhance the blood circulation and, hence, tumour accumulation. In preclinical studies, [177Lu]Lu-SibuDAB, a radiopharmaceutical with moderate albumin-binding properties, outperformed [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T. The aim of this study was to evaluate the dosimetry of [177Lu]Lu-SibuDAB in patients diagnosed mCRPC. METHODS Seventeen patients (median age 72 years, range 63‒83) diagnosed with progressive disease of mCRPC were included in this prospective study after exhausting all available treatment options. They were injected with 5.3 ± 0.5 GBq (mean ± standard deviation) [177Lu]Lu-SibuDAB as a first treatment cycle. Sixteen of these patients underwent sequential whole-body SPECT/CT and activity determination in venous blood samples for dosimetry purposes. Absorbed doses to the salivary glands, liver, spleen, kidneys, and red marrow as well as selected tumour lesions were calculated in OLINDA/EXM™ and compared to published values for previously established PSMA radiopharmaceuticals. RESULTS Absorbed dose coefficients (ADC) to tumours (9.9 ± 5.4 Gy/GBq) were about 2-fold higher than those reported for clinically approved PSMA radiopharmaceuticals. ADC to salivary glands, liver, spleen, kidneys and red marrow were higher (0.5 ± 0.2, 0.2 ± 0.05, 0.2 ± 0.1, 1.8 ± 0.6, 0.1 ± 0.04 Gy/GBq, respectively) than for [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T, but lower than for [177Lu]Lu-PSMA-ALB-56, a previously investigated long-circulating PSMA radiopharmaceutical. The tumour-to-kidneys, tumour-to-red marrow, tumour-to-salivary glands ADC ratio were 6.6, 102, 33.1. These ratios were comparable to those of [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T for kidneys and red-marrow, but higher for salivary glands. CONCLUSION [177Lu]Lu-SibuDAB showed a prolonged blood circulation time and, hence, a significantly increased absorbed tumour dose, while tumour-to-organ ADC ratios were similar to conventional PSMA radiopharmaceuticals. Further clinical investigations to evaluate the efficacy and safety of [177Lu]Lu-SibuDAB are, thus, warranted.
Collapse
Affiliation(s)
- Philipp Ritt
- ITM Oncologics GmbH, Lichtenbergstrasse 1, 85748, Garching, Munich, Germany.
- Chair for Clinical Nuclear Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| | - René Fernández
- Center for Nuclear Medicine & PET/CT Positronmed, 7501068, Providencia, Santiago, Chile
| | - Cristian Soza-Ried
- Center for Nuclear Medicine & PET/CT Positronmed, 7501068, Providencia, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Heinz Nicolai
- Center for Nuclear Medicine & PET/CT Positronmed, 7501068, Providencia, Santiago, Chile
- Departamento de Urología, Hospital Clínico San Borja Arriarán, Universidad de Chile, Santiago, Chile
| | - Horacio Amaral
- Center for Nuclear Medicine & PET/CT Positronmed, 7501068, Providencia, Santiago, Chile
- Positronpharma SA, 7501068, Providencia, Santiago, Chile
| | - Korbinian Krieger
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, 5232, Villigen-PSI, Switzerland
| | - Amanda Rotger
- ITM Oncologics GmbH, Lichtenbergstrasse 1, 85748, Garching, Munich, Germany
| | | | - Roger Schibli
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Vasko Kramer
- Center for Nuclear Medicine & PET/CT Positronmed, 7501068, Providencia, Santiago, Chile
- Positronpharma SA, 7501068, Providencia, Santiago, Chile
| |
Collapse
|
11
|
Sergeeva OV, Luo L, Guiseppi-Elie A. Cancer theragnostics: closing the loop for advanced personalized cancer treatment through the platform integration of therapeutics and diagnostics. Front Bioeng Biotechnol 2025; 12:1499474. [PMID: 39898278 PMCID: PMC11782185 DOI: 10.3389/fbioe.2024.1499474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Cancer continues to be one of the leading causes of death worldwide, and conventional cancer therapies such as chemotherapy, radiation therapy, and surgery have limitations. RNA therapy and cancer vaccines hold considerable promise as an alternative to conventional therapies for their ability to enable personalized therapy with improved efficacy and reduced side effects. The principal approach of cancer vaccines is to induce a specific immune response against cancer cells. However, a major challenge in cancer immunotherapy is to predict which patients will respond to treatment and to monitor the efficacy of the vaccine during treatment. Theragnostics, an integration of diagnostic and therapeutic capabilities into a single hybrid platform system, has the potential to address these challenges by enabling real-time monitoring of treatment response while allowing endogenously controlled personalized treatment adjustments. In this article, we review the current state-of-the-art in theragnostics for cancer vaccines and RNA therapy, including imaging agents, biomarkers, and other diagnostic tools relevant to cancer, and their application in cancer therapy development and personalization. We also discuss the opportunities and challenges for further development and clinical translation of theragnostics in cancer vaccines.
Collapse
Affiliation(s)
| | - Liang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Anthony Guiseppi-Elie
- Bioelectronics, Biosensors and Biochips (C3B), Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Full Affiliate Member, Houston Methodist Research Institute, Houston, TX, United States
- ABTECH Scientific, Inc., Biotechnology Research Park, Richmond, VA, United States
| |
Collapse
|
12
|
Li Z, Ruan Q, Jiang Y, Wang Q, Yin G, Feng J, Zhang J. Current Status and Perspectives of Novel Radiopharmaceuticals with Heterologous Dual-targeted Functions: 2013-2023. J Med Chem 2024; 67:21644-21670. [PMID: 39648432 DOI: 10.1021/acs.jmedchem.4c01608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Radiotracers provide molecular- and cellular-level information in a noninvasive manner and have become important tools for precision medicine. In particular, the successful clinical application of radioligand therapeutic (RLT) has further strengthened the role of nuclear medicine in clinical treatment. The complicated microenvironment of the lesion has rendered traditional single-targeted radiopharmaceuticals incapable of fully meeting the requirements. The design and development of dual-targeted and multitargeted radiopharmaceuticals have rapidly emerged. In recent years, significant progress has been made in the development of heterologous dual-targeted radiopharmaceuticals. This perspective aims to provide a comprehensive overview of the recent progress in these heterologous dual-targeted radiopharmaceuticals, with a special focus on the design of ligand structures, pharmacological properties, and preclinical and clinical evaluation. Furthermore, future directions are discussed from this perspective.
Collapse
Affiliation(s)
- Zuojie Li
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
13
|
Lima H, Etchebehere M, Bogoni M, Torricelli C, Nogueira-Lima E, Deflon VM, Lima M, Etchebehere E. Theranostics Nuclear Medicine in Prostate Cancer. Pharmaceuticals (Basel) 2024; 17:1483. [PMID: 39598394 PMCID: PMC11597825 DOI: 10.3390/ph17111483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Theranostic Nuclear Medicine is based on the idea of combining the same molecule (or drug) with different radioisotopes for both diagnosis and treatment, a concept that emerged in the early 1940s with the use of radioactive iodine for thyroid diseases. Theranostic Nuclear Medicine has since expanded to diseases of higher incidence, such as prostate cancer, with several imaging methods used to assess the extent of the disease and the corresponding radiopharmaceuticals used for treatment. For example, by detecting osteoblastic metastases by bone scintigraphy, corresponding radiopharmaceuticals with therapeutic properties can be administered to eliminate or reduce pain associated with metastases and/or determine overall survival gain. The purpose of this review is to discuss the role of Theranostic Nuclear Medicine in prostate cancer, addressing the main diagnostic imaging studies with their corresponding treatments in the Theranostic model.
Collapse
Affiliation(s)
- Helena Lima
- Faculdade de Medicina, Pontifícia Universidade Católica de Campinas (PUCC), Campinas 13087-571, Brazil;
| | - Marina Etchebehere
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil;
| | - Mateos Bogoni
- Hospital Erasto Gaertner, Curitiba 81520-060, Brazil;
- Diagnóstico Avançado por Imagem (DAPI), Curitiba 80430-210, Brazil
| | - Caroline Torricelli
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
| | - Ellen Nogueira-Lima
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
| | - Victor M. Deflon
- São Carlos Institute of Chemistry, University of São Paulo (USP), São Carlos 13566-590, Brazil;
| | - Mariana Lima
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
- Medicina Nuclear de Campinas (Grupo MND), Campinas 13020-432, Brazil
| | - Elba Etchebehere
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
- Medicina Nuclear de Campinas (Grupo MND), Campinas 13020-432, Brazil
| |
Collapse
|
14
|
Huangfu Z, Yang J, Sun J, Xu B, Tao L, Wu J, Wang F, Wang G, Meng F, Zhong Z. PSMA and Sigma-1 receptor dual-targeted peptide mediates superior radionuclide imaging and therapy of prostate cancer. J Control Release 2024; 375:767-775. [PMID: 39332777 DOI: 10.1016/j.jconrel.2024.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/26/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Radionuclide therapy, in particular peptide receptor radionuclide therapy (PRRT), has emerged as a valuable means to combat malignant tumors. The specific affinity of ACUPA peptide toward prostate-specific membrane antigen (PSMA) renders the successful development of PRRT for prostate cancer. The clinical outcome of PRRT is, however, generally challenged by moderate tumor uptake and off-target toxicity. Here, we report on a novel design of Sigma-1 receptor and PSMA dual-receptor targeted peptide (S1R/PSMA-P) for superior radionuclide imaging and therapy of prostate cancer. S1R/PSMA-P was acquired with good purity and could efficiently be labeled with 177Lu to yield 177Lu-S1R/PSMA-P with high specific activity and radiostability. Interestingly, 177Lu-S1R/PSMA-P revealed greatly enhanced affinity to LNCaP cells over single-targeted control 177Lu-PSMA-617. The single photon emission computed tomography (SPECT) imaging demonstrated exceptional uptake and retention of 177Lu-S1R/PSMA-P in LNCaP tumor, affording about 2-fold better tumor accumulation while largely reduced uptake by most normal tissues compared to 177Lu-PSMA-617. The selective uptake in LNCaP tumor was also visualized by positron emission tomography (PET) with 68Ga-S1R/PSMA-P. In accordance, a single and low dosage of 177Lu-S1R/PSMA-P at 11.1 MBq effectively suppressed tumor growth without causing apparent side effects. This dual-targeting strategy presents an appealing radionuclide therapy for malignant tumors.
Collapse
Affiliation(s)
- Zhenyuan Huangfu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Juan Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Bin Xu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Lei Tao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiang Wu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, People's Republic of China.
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
15
|
Bai L, Sun P, Huang S, Shi D, Cheng K, Cai Z, Dong Y, Tang G. Comparative study of [ 18F]AlF-PAI-PDL1p and [ 68Ga]Ga-PAI-PDL1p as novel PD-L1 targeting PET probes for tumor imaging. Bioorg Chem 2024; 151:107660. [PMID: 39079391 DOI: 10.1016/j.bioorg.2024.107660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 08/30/2024]
Abstract
PD-L1 is expressed in many tumors but rarely in normal tissues, therefore, it can be a target of PET imaging. In this work, we developed new peptide-based PET probes [18F]AlF-PAI-PDL1p and [68Ga]Ga-PAI-PDL1p with yields of 20-25 % and 40-55 %, respectively. [18F]AlF-PAI-PDL1p and [68Ga]Ga-PAI-PDL1p were synthesized within 30 min with high molar activities. [18F]AlF-PAI-PDL1p and [68Ga]Ga-PAI-PDL1p showed good stability in vivo and in vitro. In vitro cell studies showed [18F]AlF-PAI-PDL1p and [68Ga]Ga-PAI-PDL1p target PD-L1 specifically, with high uptake of 61.52 ± 4.39 and 19.29 ± 2.17 %ID/1 million cells in B16F10 cells at 60 min, respectively. Biodistribution results showed that both [18F]AlF-PAI-PDL1p and [68Ga]Ga-PAI-PDL1p had lower liver accumulation. In vivo PET imaging results showed that [18F]AlF-PAI-PDL1p had a high tumor uptake of 4.23 ± 0.81 %ID/g at 2 h and increased uptake of 6.60 ± 1.01 %ID/g at 12 h. [68Ga]Ga-PAI-PDL1p also showed high tumor uptake of 2.30 ± 0.20 %ID/g at 2 h and slightly increased uptake of 3.80 ± 0.26 %ID/g at 6 h. In conclusion, [18F]AlF-PAI-PDL1p and [68Ga]Ga-PAI-PDL1 seemed to be potential tracers for PET imaging of PD-L1 expression.
Collapse
Affiliation(s)
- Lu Bai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Penghui Sun
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Shun Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Dazhi Shi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhikai Cai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Ye Dong
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Ganghua Tang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| |
Collapse
|
16
|
Abdollahi H, Fele-Paranj A, Rahmim A. Model-Informed Radiopharmaceutical Therapy Optimization: A Study on the Impact of PBPK Model Parameters on Physical, Biological, and Statistical Measures in 177Lu-PSMA Therapy. Cancers (Basel) 2024; 16:3120. [PMID: 39335092 PMCID: PMC11430653 DOI: 10.3390/cancers16183120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Purpose: To investigate the impact of physiologically based pharmacokinetic (PBPK) parameters on physical, biological, and statistical measures in lutetium-177-labeled radiopharmaceutical therapies (RPTs) targeting the prostate-specific membrane antigen (PSMA). Methods: Using a clinically validated PBPK model, realistic time-activity curves (TACs) for tumors, salivary glands, and kidneys were generated based on various model parameters. These TACs were used to calculate the area-under-the-TAC (AUC), dose, biologically effective dose (BED), and figure-of-merit BED (fBED). The effects of these parameters on radiobiological, pharmacokinetic, time, and statistical features were assessed. Results: Manipulating PBPK parameters significantly influenced AUC, dose, BED, and fBED outcomes across four different BED models. Higher association rates increased AUC, dose, and BED values for tumors, with minimal impact on non-target organs. Increased internalization rates reduced AUC and dose for tumors and kidneys. Higher serum protein-binding rates decreased AUC and dose for all tissues. Elevated tumor receptor density and ligand amounts enhanced uptake and effectiveness in tumors. Larger tumor volumes required dosimetry adjustments to maintain efficacy. Setting the tumor release rate to zero intensified the impact of association and internalization rates, enhancing tumor targeting while minimizing the effects on salivary glands and kidneys. Conclusions: Optimizing PBPK parameters can enhance the efficacy of lutetium-177-labeled RPTs targeting PSMA, providing insights for personalized and effective treatment regimens to minimize toxicity and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
| | - Ali Fele-Paranj
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
- Department of Mathematics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
- Department of Physics & Astronomy, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
17
|
Qiao Z, Xu J, Gallazzi F, Fisher DR, Gonzalez R, Kwak J, Miao Y. Effect of Ibuprofen as an Albumin Binder on Melanoma-Targeting Properties of 177Lu-Labeled Ibuprofen-Conjugated Alpha-Melanocyte-Stimulating Hormone Peptides. Mol Pharm 2024; 21:4004-4011. [PMID: 38973113 DOI: 10.1021/acs.molpharmaceut.4c00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
The purpose of this study was to examine how the introduction of ibuprofen (IBU) affected tumor-targeting and biodistribution properties of 177Lu-labeled IBU-conjugated alpha-melanocyte-stimulating hormone peptides. The IBU was used as an albumin binder and conjugated to the DOTA-Lys moiety without or with a linker to yield DOTA-Lys(IBU)-GG-Nle-CycMSHhex {1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-Lys(IBU)-Gly-Gly-Nle-c[Asp-His-DPhe-Arg-Trp-Lys]-CONH2}, DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex, DOTA-Lys(Asn-IBU)-GGNle-CycMSHhex, and DOTA-Lys(Dab-IBU)-GGNle-CycMSHhex peptides. Their melanocortin-receptor 1 (MC1R) binding affinities were determined on B16/F10 melanoma cells first. Then the biodistribution of 177Lu-labeled peptides was determined on B16/F10 melanoma-bearing C57 mice at 2 h postinjection to choose the lead peptide for further examination. The full biodistribution and melanoma imaging properties of 177Lu-DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex were further evaluated using B16/F10 melanoma-bearing C57 mice. DOTA-Lys(IBU)-GG-Nle-CycMSHhex, DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex, DOTA-Lys(Asn-IBU)-GGNle-CycMSHhex, and DOTA-Lys(Dab-IBU)-GGNle-CycMSHhex displayed the IC50 values of 1.41 ± 0.37, 1.52 ± 0.08, 0.03 ± 0.01, and 0.58 ± 0.06 nM on B16/F10 melanoma cells, respectively. 177Lu-DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex exhibited the lowest liver and kidney uptake among all four designed 177Lu peptides. Therefore, 177Lu-DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex was further evaluated for its full biodistribution and melanoma imaging properties. The B16/F10 melanoma uptake of 177Lu-DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex was 19.5 ± 3.12, 24.12 ± 3.35, 23.85 ± 2.08, and 10.80 ± 2.89% ID/g at 0.5, 2, 4, and 24 h postinjection, respectively. Moreover, 177Lu-DOTA-Lys(Asp-IBU)-GGNle-CycMSHhex could clearly visualize the B16/F10 melanoma lesions at 2 h postinjection. The conjugation of IBU with or without a linker to GGNle-CycMSHhex affected the MC1R binding affinities of the designed peptides. The charge of the linker played a key role in the liver and kidney uptake of 177Lu-Asp-IBU, 177Lu-Asn-IBU, and 177Lu-Dab-IBU. 177Lu-Asp-IBU exhibited higher tumor/liver and tumor/kidney uptake ratios than those of 177Lu-Asn-IBU and 177Lu-Dab-IBU, underscoring its potential evaluation for melanoma therapy in the future.
Collapse
Affiliation(s)
- Zheng Qiao
- Department of Radiology, University of Colorado Denver, Aurora, Colorado 80045, United States
| | - Jingli Xu
- Department of Radiology, University of Colorado Denver, Aurora, Colorado 80045, United States
| | - Fabio Gallazzi
- Department of Chemistry and Molecular Interactions Core, University of Missouri, Columbia, Missouri 65211, United States
| | - Darrell R Fisher
- Versant Medical Physics and Radiation Safety, Richland, Washington 99354, United States
| | - Rene Gonzalez
- Department of Medical Oncology, University of Colorado Denver, Aurora, Colorado 80045, United States
| | - Jennifer Kwak
- Department of Radiology, University of Colorado Denver, Aurora, Colorado 80045, United States
| | - Yubin Miao
- Department of Radiology, University of Colorado Denver, Aurora, Colorado 80045, United States
| |
Collapse
|
18
|
Lindeman SD, Booth OC, Tudi P, Schleinkofer TC, Moss JN, Kearney NB, Mukkamala R, Thompson LK, Modany MA, Srinivasarao M, Low PS. FAP Radioligand Linker Optimization Improves Tumor Dose and Tumor-to-Healthy Organ Ratios in 4T1 Syngeneic Model. J Med Chem 2024; 67:11827-11840. [PMID: 39013156 DOI: 10.1021/acs.jmedchem.4c00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Fibroblast activation protein (FAP) has attracted considerable attention as a possible target for the radiotherapy of solid tumors. Unfortunately, initial efforts to treat solid tumors with FAP-targeted radionuclides have yielded only modest clinical responses, suggesting that further improvements in the molecular design of FAP-targeted radiopharmaceutical therapies (RPT) are warranted. In this study, we report several advances on the previously described FAP6 radioligand that increase tumor retention and accelerate healthy tissue clearance. Seven FAP6 derivatives with different linkers or albumin binders were synthesized, radiolabeled, and investigated for their effects on binding and cellular uptake. The radioligands were then characterized in 4T1 tumor-bearing Balb/c mice using both single-photon emission computed tomography (SPECT) and ex vivo biodistribution analyses to identify the conjugate with the best tumor retention and tumor-to-healthy organ ratios. The results reveal an optimized FAP6 radioligand that exhibits efficacy and safety properties that potentially justify its translation into the clinic.
Collapse
Affiliation(s)
- Spencer D Lindeman
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- MorphImmune, Inc., 1281 Win Hentschel Blvd, West Lafayette, Indiana 47906, United States
| | - Owen C Booth
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pooja Tudi
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Taylor C Schleinkofer
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jackson N Moss
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Nicholas B Kearney
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ramesh Mukkamala
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lauren K Thompson
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mollie A Modany
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- MorphImmune, Inc., 1281 Win Hentschel Blvd, West Lafayette, Indiana 47906, United States
| |
Collapse
|
19
|
Li L, Wang J, Wang G, Wang R, Jin W, Zang J, Sui H, Jia C, Jiang Y, Hong H, Zhu L, Alexoff D, Ploessl K, Kung HF, Zhu Z. Comparison of novel PSMA-targeting [ 177Lu]Lu-P17-087 with its albumin binding derivative [ 177Lu]Lu-P17-088 in metastatic castration-resistant prostate cancer patients: a first-in-human study. Eur J Nucl Med Mol Imaging 2024; 51:2794-2805. [PMID: 38658392 DOI: 10.1007/s00259-024-06721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is a promising target for diagnosis and radioligand therapy (RLT) of prostate cancer. Two novel PSMA-targeting radionuclide therapy agents, [177Lu]Lu-P17-087, and its albumin binder modified derivative, [177Lu]Lu-P17-088, were evaluated in metastatic castration-resistant prostate cancer (mCRPC) patients. The primary endpoint was dosimetry evaluation, the second endpoint was radiation toxicity assessment (CTCAE 5.0) and PSA response (PCWG3). METHODS Patients with PSMA-positive tumors were enrolled after [68Ga]Ga-PSMA-11 PET/CT scan. Five mCRPC patients received [177Lu]Lu-P17-087 and four other patients received [177Lu]Lu-P17-088 (1.2 GBq/patient). Multiple whole body planar scintigraphy was performed at 1.5, 4, 24, 48, 72, 120 and 168 h after injection and one SPECT/CT imaging was performed at 24 h post-injection for each patient. Dosimetry evaluation was compared in both patient groups. RESULTS Patients showed no major clinical side-effects under this low dose treatment. As expected [177Lu]Lu-P17-088 with longer blood circulation (due to its albumin binding) exhibited higher effective doses than [177Lu]Lu-P17-087 (0.151 ± 0.036 vs. 0.056 ± 0.019 mGy/MBq, P = 0.001). Similarly, red marrow received 0.119 ± 0.068 and 0.048 ± 0.020 mGy/MBq, while kidney doses were 0.119 ± 0.068 and 0.046 ± 0.022 mGy/MBq, respectively. [177Lu]Lu-P17-087 demonstrated excellent tumor uptake and faster kinetics; while [177Lu]Lu-P17-088 displayed a slower washout and higher average dose (7.75 ± 4.18 vs. 4.72 ± 2.29 mGy/MBq, P = 0.018). After administration of [177Lu]Lu-P17-087 and [177Lu]Lu-P17-088, 3/5 and 3/4 patients showed reducing PSA values, respectively. CONCLUSION [177Lu]Lu-P17-088 and [177Lu]Lu-P17-087 displayed different pharmacokinetics but excellent PSMA-targeting dose delivery in mCRPC patients. These two agents are promising RLT agents for personalized treatment of mCRPC. Further studies with increased dose and frequency of RLT are warranted to evaluate the potential therapeutic efficacy. TRIAL REGISTRATION 177Lu-P17-087/177Lu-P17-088 in Patients with Metastatic Castration-resistant Prostate Cancer (NCT05603559, Registered at 25 October, 2022). URL OF REGISTRY: https://classic. CLINICALTRIALS gov/ct2/show/NCT05603559 .
Collapse
Affiliation(s)
- Linlin Li
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China
| | - Jiarou Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China
| | - Guochang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Rongxi Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China
| | - Wenbin Jin
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China
| | - Jie Zang
- Department of Nuclear Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Huimin Sui
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China
| | - Chenhao Jia
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China
| | - Yuanyuan Jiang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China
| | - Haiyan Hong
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China
| | - Lin Zhu
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China
| | - David Alexoff
- Five Eleven Pharma Inc, Philadelphia, PA, 19104, USA
| | - Karl Ploessl
- Five Eleven Pharma Inc, Philadelphia, PA, 19104, USA
| | - Hank F Kung
- Five Eleven Pharma Inc, Philadelphia, PA, 19104, USA.
- Department of Radiology, University of Pennsylvania, 3700 Market Street, Room 305, Philadelphia, PA, 19104, USA.
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing St., Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
20
|
Mo C, Sun P, Liang H, Chen Z, Wang M, Fu L, Huang S, Tang G. Synthesis and preclinical evaluation of a novel probe [ 18F]AlF-NOTA-IPB-GPC3P for PET imaging of GPC3 positive tumor. Bioorg Chem 2024; 147:107352. [PMID: 38640719 DOI: 10.1016/j.bioorg.2024.107352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/23/2024] [Accepted: 04/05/2024] [Indexed: 04/21/2024]
Abstract
Glypican-3 (GPC3) is markedly overexpressed in hepatocellular carcinoma (HCC) and not expressed in normal liver tissues. In this study, a novel peptide PET imaging agent ([18F]AlF-NOTA-IPB-GPC3P) was developed to target GPC3 expressed in tumors. The overall radiochemical yield of [18F]AlF-NOTA-IPB-GPC3P was 10-15 %, and its lipophilicity, expressed as the logD value at a pH of 7.4, was -1.18 ± 0.06 (n = 3). Compared to the previously reported tracer [18F]AlF-GP2633, [18F]AlF-NOTA-IPB-GPC3P exhibited higher cellular uptake (15.13 vs 5.96) and internalized rate (80.63 % vs 35.93 %) in Huh7 cells at 120 min. Micro-PET/CT and biodistribution studies further demonstrated that [18F]AlF-NOTA-IPB-GPC3P exhibited significantly increased tumor uptake and prolonged tumor residence in Huh7 tumors compared to [18F]AlF-GP2633 (4.66 ± 0.22 % ID/g vs 0.72 ± 0.09 % ID/g at 60 min, p < 0.001; 5.05 ± 0.23 % ID/g vs 0.35 ± 0.08 % ID/g at 120 min, p < 0.001, respectively). Furthermore, the tumor-to-organ ratios of [18F]AlF-NOTA-IPB-GPC3P surpassed those of [18F]AlF-GP2633. Our results support the utilization of [18F]AlF-NOTA-IPB-GPC3P as a PET imaging agent targeting the GPC3 receptor for tumor detection.
Collapse
Affiliation(s)
- Chunwei Mo
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China
| | - Penghui Sun
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China
| | - Haoran Liang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China
| | - Zihao Chen
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China
| | - Meng Wang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China
| | - Lilan Fu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China
| | - Shun Huang
- Department of Nuclear Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China.
| | - Ganghua Tang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
21
|
Tsuchihashi S, Nakashima K, Watanabe H, Ono M. Synthesis and evaluation of novel trifunctional chelating agents for pretargeting approach using albumin binder to improve tumor accumulation. Nucl Med Biol 2024; 132-133:108911. [PMID: 38614036 DOI: 10.1016/j.nucmedbio.2024.108911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION The pretargeting approach consists of in vivo ligation between pre-injected antibodies and low-molecular-weight radiolabeled effectors. The advantage of the pretargeting approach is to improve a tumor-to-background ratio, but the disadvantage is to compromise tumor accumulation. In this study, we applied albumin binder (ALB) to the pretargeting approach to overcome low tumor accumulation. METHODS We synthesized two novel trifunctional effectors containing an ALB moiety, a chelator, and a different tetrazine and two corresponding effectors without an ALB moiety. Albumin-binding assays and stability assays were performed using 111In-labeled effectors. Measurements of reaction rate constant were conducted using 111In-labeled effectors and anti-HER2 antibody trastuzumab modified by trans-cyclooctene, which drives the click reaction with tetrazine. Biodistribution studies using HER2-expressing tumor-bearing mice were performed with or without the pretargeting approach. RESULTS In albumin-binding assays, ALB-containing effectors exhibited a marked binding to albumin. Two ALB-containing effectors showed the difference in the reactivity and the slight difference in the stability. In biodistribution studies without the pretargeting approach, two ALB-containing effectors showed different pharmacokinetics in blood retention. With the pretargeting approach, the tumor accumulation was improved by the introduction of ALB and the highest tumor accumulation was observed in using the ALB-containing effector with higher blood retention. CONCLUSION These results suggest that the application of ALB to the pretargeting approach is effective to improve tumor accumulation, and the structure of tetrazine influences the utility of ALB-containing effectors.
Collapse
Affiliation(s)
- Shohei Tsuchihashi
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
22
|
Vaccarin C, Mapanao AK, Deberle LM, Becker AE, Borgna F, Marzaro G, Schibli R, Müller C. Design and Preclinical Evaluation of a Novel Prostate-Specific Membrane Antigen Radioligand Modified with a Transthyretin Binder. Cancers (Basel) 2024; 16:1262. [PMID: 38610940 PMCID: PMC11011029 DOI: 10.3390/cancers16071262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Transthyretin binders have previously been used to improve the pharmacokinetic properties of small-molecule drug conjugates and could, thus, be utilized for radiopharmaceuticals as an alternative to the widely explored "albumin binder concept". In this study, a novel PSMA ligand modified with a transthyretin-binding entity (TB-01) was synthesized and labeled with lutetium-177 to obtain [177Lu]Lu-PSMA-TB-01. A high and specific uptake of [177Lu]Lu-PSMA-TB-01 was found in PSMA-positive PC-3 PIP cells (69 ± 3% after 4 h incubation), while uptake in PSMA-negative PC-3 flu cells was negligible (<1%). In vitro binding studies showed a 174-fold stronger affinity of [177Lu]Lu-PSMA-TB-01 to transthyretin than to human serum albumin. Biodistribution studies in PC-3 PIP/flu tumor-bearing mice confirmed the enhanced blood retention of [177Lu]Lu-PSMA-TB-01 (16 ± 1% IA/g at 1 h p.i.), which translated to a high tumor uptake (69 ± 13% IA/g at 4 h p.i.) with only slow wash-out over time (31 ± 8% IA/g at 96 h p.i.), while accumulation in the PC-3 flu tumor and non-targeted normal tissue was reasonably low. Further optimization of the radioligand design would be necessary to fine-tune the biodistribution and enable its use for therapeutic purposes. This study was the first of this kind and could motivate the use of the "transthyretin binder concept" for the development of future radiopharmaceuticals.
Collapse
Affiliation(s)
- Christian Vaccarin
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
| | - Luisa M. Deberle
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
| | - Anna E. Becker
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
| | - Francesca Borgna
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, I-35131 Padua, Italy;
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (C.V.); (A.K.M.); (L.M.D.); (A.E.B.); (F.B.); (R.S.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
23
|
Holzleitner N, Fischer S, Maniyankerikalam I, Beck R, Lapa C, Wester HJ, Günther T. Significant reduction of activity retention in the kidneys via optimized linker sequences in radiohybrid-based minigastrin analogs. EJNMMI Res 2024; 14:23. [PMID: 38429609 PMCID: PMC10907560 DOI: 10.1186/s13550-024-01087-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND We recently introduced radiohybrid (rh)-based minigastrin analogs e.g., DOTA-rhCCK-18 (DOTA-D-Dap(p-SiFA)-(D-γ-Glu)8-Ala-Tyr-Gly-Trp-Nle-Asp-Phe-NH2), that revealed substantially increased activity retention in the tumor. However, one major drawback of these first generation rh-based cholecystokinin-2 receptor (CCK-2R) ligands is their elevated activity levels in the kidneys, especially at later time points (24 h p.i.). Therefore, this study aimed to reduce kidney retention with regard to a therapeutic use via substitution of negatively charged D-glutamic acid moieties by hydrophilic uncharged polyethylene glycol (PEG) linkers of various length ((PEG)4 to (PEG)11). Furthermore, the influence of differently charged silicon-based fluoride acceptor (SiFA)-moieties (p-SiFA: neutral, SiFA-ipa: negatively charged, and SiFAlin: positively charged) on in vitro properties of minigastrin analogs was evaluated. Out of all compounds evaluated in vitro, the two most promising minigastrin analogs were further investigated in vivo. RESULTS CCK-2R affinity of most compounds evaluated was found to be in a range of 8-20 nM (by means of apparent IC50), while ligands containing a SiFA-ipa moiety displayed elevated IC50 values. Lipophilicity was noticeably lower for compounds containing a D-γ-glutamate (D-γ-Glu) moiety next to the D-Dap(SiFA) unit as compared to their counterparts lacking the additional negative charge. Within this study, combining the most favorable CCK-2R affinity and lipophilicity, [177/natLu]Lu-DOTA-rhCCK-70 (DOTA-D-Dap(p-SiFA)-D-γ-Glu-(PEG)7-D-γ-Glu-(PEG)3-Trp-(N-Me)Nle-Asp-1-Nal-NH2; IC50: 12.6 ± 2.0 nM; logD7.4: - 1.67 ± 0.08) and [177/natLu]Lu-DOTA-rhCCK-91 (DOTA-D-Dap(SiFAlin)-D-γ-Glu-(PEG)4-D-γ-Glu-(PEG)3-Trp-(N-Me)Nle-Asp-1-Nal-NH2; IC50: 8.6 ± 0.7 nM; logD7.4 = - 1.66 ± 0.07) were further evaluated in vivo. Biodistribution data of both compounds revealed significantly reduced (p < 0.0001) activity accumulation in the kidneys compared to [177Lu]Lu-DOTA-rhCCK-18 at 24 h p.i., leading to enhanced tumor-to-kidney ratios despite lower tumor uptake. However, overall tumor-to-background ratios of the novel compounds were lower than those of [177Lu]Lu-DOTA-rhCCK-18. CONCLUSION We could show that the reduction of negative charges within the linker section of radiohybrid-based minigastrin analogs led to decreased activity levels in the kidneys at 24 h p.i., while maintaining a good tumor uptake. Thus, favorable tumor-to-kidney ratios were accomplished in vivo. However, further optimization has to be done in order to improve tumor retention and general biodistribution profile.
Collapse
Affiliation(s)
- Nadine Holzleitner
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany.
| | - Sebastian Fischer
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Isabel Maniyankerikalam
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Roswitha Beck
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Bavaria, Germany
| | - Hans-Jürgen Wester
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Thomas Günther
- TUM School of Natural Sciences, Department of Chemistry, Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany.
| |
Collapse
|
24
|
de Roode KE, Joosten L, Behe M. Towards the Magic Radioactive Bullet: Improving Targeted Radionuclide Therapy by Reducing the Renal Retention of Radioligands. Pharmaceuticals (Basel) 2024; 17:256. [PMID: 38399470 PMCID: PMC10892921 DOI: 10.3390/ph17020256] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Targeted radionuclide therapy (TRT) is an emerging field and has the potential to become a major pillar in effective cancer treatment. Several pharmaceuticals are already in routine use for treating cancer, and there is still a high potential for new compounds for this application. But, a major issue for many radiolabeled low-to-moderate-molecular-weight molecules is their clearance via the kidneys and their subsequent reuptake. High renal accumulation of radioactive compounds may lead to nephrotoxicity, and therefore, the kidneys are often the dose-limiting organs in TRT with these radioligands. Over the years, different strategies have been developed aiming for reduced kidney retention and enhanced therapeutic efficacy of radioligands. In this review, we will give an overview of the efforts and achievements of the used strategies, with focus on the therapeutic potential of low-to-moderate-molecular-weight molecules. Among the strategies discussed here is coadministration of compounds that compete for binding to the endocytic receptors in the proximal tubuli. In addition, the influence of altering the molecular design of radiolabeled ligands on pharmacokinetics is discussed, which includes changes in their physicochemical properties and implementation of cleavable linkers or albumin-binding moieties. Furthermore, we discuss the influence of chelator and radionuclide choice on reabsorption of radioligands by the kidneys.
Collapse
Affiliation(s)
- Kim E. de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen, Switzerland
| |
Collapse
|
25
|
Dai R, Cai Z, Hu R, Huang Y, Fu L, Yang J, Hu K, Li H. 177Lu-Labeled Bivalent Ligands of Prostate-Specific Membrane Antigen for Endoradiotherapy of Prostate Cancer. Mol Pharm 2024; 21:883-894. [PMID: 38155100 DOI: 10.1021/acs.molpharmaceut.3c00987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2023]
Abstract
Recently, we developed a bivalent prostate-specific membrane antigen (PSMA) radioligand ([18F]AlF-Bi-PSMA), which showed higher tumor uptake and retention in PSMA-positive mouse models than the clinically used radioligands, [68Ga]Ga-PSMA-11 and [18F]PSMA-1007. Here, we developed two 177Lu-labeled bivalent PSMA ligands with (DOTA-Alb-Bi-PSMA) or without an albumin-binding motif (DOTA-Bi-PSMA) to enhance radiotherapeutic efficacy with minimal toxicity. The results demonstrated that both 177Lu-labeled bivalent radioligands showed good stability, high binding affinity, and PSMA-targeting specificity in vitro. Compared with [177Lu]Lu-PSMA-617, both [177Lu]Lu-Bi-PSMA and [177Lu]Lu-Alb-Bi-PSMA showed a higher area under the curve (AUC) of tumor accumulation and superior therapeutic efficacy. However, [177Lu]Lu-Alb-Bi-PSMA exhibited a dose-dependent increase in acute damage to kidneys. In terms of the radionuclide therapy efficacy and side effects, [177Lu]Lu-Bi-PSMA exhibited well-balanced action with high tumor-to-organs AUC ratios, resulting in remarkable therapeutic efficacy and negligible side effects. These promising results warrant further investigations to achieve the clinical translation of [177Lu]Lu-Bi-PSMA.
Collapse
Affiliation(s)
- Ruoxue Dai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Zhikai Cai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Rui Hu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yueqi Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Lilan Fu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jiaqi Yang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Kongzhen Hu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Hongsheng Li
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
26
|
Ha S, O JH, Park C, Boo SH, Yoo IR, Moon HW, Chi DY, Lee JY. Dosimetric Analysis of a Phase I Study of PSMA-Targeting Radiopharmaceutical Therapy With [ 177Lu]Ludotadipep in Patients With Metastatic Castration-Resistant Prostate Cancer. Korean J Radiol 2024; 25:179-188. [PMID: 38288897 PMCID: PMC10831299 DOI: 10.3348/kjr.2023.0656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 02/01/2024] Open
Abstract
OBJECTIVE 177Lutetium [Lu] Ludotadipep is a novel prostate-specific membrane antigen targeting therapeutic agent with an albumin motif added to increase uptake in the tumors. We assessed the biodistribution and dosimetry of [177Lu]Ludotadipep in patients with metastatic castration-resistant prostate cancer (mCRPC). MATERIALS AND METHODS Data from 25 patients (median age, 73 years; range, 60-90) with mCRPC from a phase I study with activity escalation design of single administration of [177Lu]Ludotadipep (1.85, 2.78, 3.70, 4.63, and 5.55 GBq) were assessed. Activity in the salivary glands, lungs, liver, kidneys, and spleen was estimated from whole-body scan and abdominal SPECT/CT images acquired at 2, 24, 48, 72, and 168 h after administration of [177Lu]Ludotadipep. Red marrow activity was calculated from blood samples obtained at 3, 10, 30, 60, and 180 min, and at 24, 48, and 72 h after administration. Organ- and tumor-based absorbed dose calculations were performed using IDAC-Dose 2.1. RESULTS Absorbed dose coefficient (mean ± standard deviation) of normal organs was 1.17 ± 0.81 Gy/GBq for salivary glands, 0.05 ± 0.02 Gy/GBq for lungs, 0.14 ± 0.06 Gy/GBq for liver, 0.77 ± 0.28 Gy/GBq for kidneys, 0.12 ± 0.06 Gy/GBq for spleen, and 0.07 ± 0.02 Gy/GBq for red marrow. The absorbed dose coefficient of the tumors was 10.43 ± 7.77 Gy/GBq. CONCLUSION [177Lu]Ludotadipep is expected to be safe at the dose of 3.7 GBq times 6 cycles planned for a phase II clinical trial with kidneys and bone marrow being the critical organs, and shows a high tumor absorbed dose.
Collapse
Affiliation(s)
- Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joo Hyun O
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Chansoo Park
- Research Institute of Labeling, FutureChem Co., Ltd., Seoul, Republic of Korea
| | - Sun Ha Boo
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ie Ryung Yoo
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyong Woo Moon
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dae Yoon Chi
- Research Institute of Labeling, FutureChem Co., Ltd., Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Morgan KA, Rudd SE, Noor A, Donnelly PS. Theranostic Nuclear Medicine with Gallium-68, Lutetium-177, Copper-64/67, Actinium-225, and Lead-212/203 Radionuclides. Chem Rev 2023; 123:12004-12035. [PMID: 37796539 DOI: 10.1021/acs.chemrev.3c00456] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Molecular changes in malignant tissue can lead to an increase in the expression levels of various proteins or receptors that can be used to target the disease. In oncology, diagnostic imaging and radiotherapy of tumors is possible by attaching an appropriate radionuclide to molecules that selectively bind to these target proteins. The term "theranostics" describes the use of a diagnostic tool to predict the efficacy of a therapeutic option. Molecules radiolabeled with γ-emitting or β+-emitting radionuclides can be used for diagnostic imaging using single photon emission computed tomography or positron emission tomography. Radionuclide therapy of disease sites is possible with either α-, β-, or Auger-emitting radionuclides that induce irreversible damage to DNA. This Focus Review centers on the chemistry of theranostic approaches using metal radionuclides for imaging and therapy. The use of tracers that contain β+-emitting gallium-68 and β-emitting lutetium-177 will be discussed in the context of agents in clinical use for the diagnostic imaging and therapy of neuroendocrine tumors and prostate cancer. A particular emphasis is then placed on the chemistry involved in the development of theranostic approaches that use copper-64 for imaging and copper-67 for therapy with functionalized sarcophagine cage amine ligands. Targeted therapy with radionuclides that emit α particles has potential to be of particular use in late-stage disease where there are limited options, and the role of actinium-225 and lead-212 in this area is also discussed. Finally, we highlight the challenges that impede further adoption of radiotheranostic concepts while highlighting exciting opportunities and prospects.
Collapse
Affiliation(s)
- Katherine A Morgan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| |
Collapse
|
28
|
Tschan VJ, Busslinger SD, Bernhardt P, Grundler PV, Zeevaart JR, Köster U, van der Meulen NP, Schibli R, Müller C. Albumin-Binding and Conventional PSMA Ligands in Combination with 161Tb: Biodistribution, Dosimetry, and Preclinical Therapy. J Nucl Med 2023; 64:1625-1631. [PMID: 37442604 DOI: 10.2967/jnumed.123.265524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
The favorable decay characteristics of 161Tb attracted the interest of clinicians in using this novel radionuclide for radioligand therapy (RLT). 161Tb decays with a similar half-life to 177Lu, but beyond the emission of β--particles and γ-rays, 161Tb also emits conversion and Auger electrons, which may be particularly effective to eliminate micrometastases. The aim of this study was to compare the dosimetry and therapeutic efficacy of 161Tb and 177Lu in tumor-bearing mice using SibuDAB and PSMA-I&T, which differ in their blood residence time and tumor uptake. Methods: [161Tb]Tb-SibuDAB and [161Tb]Tb-PSMA-I&T were evaluated in vitro and investigated in biodistribution, imaging, and therapy studies using PC-3 PIP tumor-bearing mice. The 177Lu-labeled counterparts served for dose calculations and comparison of therapeutic efficacy. The tolerability of RLT in mice was monitored on the basis of body mass, blood plasma parameters, blood cell counts, and the histology of relevant organs and tissues. Results: The prostate-specific membrane antigen (PSMA)-targeting radioligands, irrespective of whether labeled with 161Tb or 177Lu, showed similar in vitro data and comparable tissue distribution profiles. As a result of the albumin-binding properties, [161Tb]Tb/[177Lu]Lu-SibuDAB had an enhanced blood residence time and higher tumor uptake (62%-69% injected activity per gram at 24 h after injection) than [161Tb]Tb/[177Lu]Lu-PSMA-I&T (30%-35% injected activity per gram at 24 h after injection). [161Tb]Tb-SibuDAB inhibited tumor growth more effectively than [161Tb]Tb-PSMA-I&T, as can be ascribed to its 4-fold increased absorbed tumor dose. At any of the applied activities, the 161Tb-based radioligands were therapeutically more effective than their 177Lu-labeled counterparts, as agreed with the approximately 40% increased tumor dose of 161Tb compared with that of 177Lu. Under the given experimental conditions, no obvious adverse events were observed. Conclusion: The data of this study indicate the promising potential of 161Tb in combination with SibuDAB for RLT of prostate cancer. Future clinical studies using 161Tb-based RLT will shed light on a potential clinical benefit of 161Tb over 177Lu.
Collapse
Affiliation(s)
- Viviane J Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Peter Bernhardt
- Department of Radiation Physics, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, South Africa
| | | | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, Switzerland; and
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland;
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Szücs D, Szabó JP, Arató V, Gyuricza B, Szikra D, Tóth I, Képes Z, Trencsényi G, Fekete A. Investigation of the Effect on the Albumin Binding Moiety for the Pharmacokinetic Properties of 68Ga-, 205/206Bi-, and 177Lu-Labeled NAPamide-Based Radiopharmaceuticals. Pharmaceuticals (Basel) 2023; 16:1280. [PMID: 37765089 PMCID: PMC10536547 DOI: 10.3390/ph16091280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Although radiolabeled alpha-melanocyte stimulating hormone-analogue NAPamide derivatives are valuable melanoma-specific diagnostic probes, their rapid elimination kinetics and high renal uptake may preclude them from being used in clinical settings. We aimed at improving the pharmacokinetics of radiolabeled DOTA-NAPamide compounds by incorporating a 4-(p-iodo-phenyl)-butanoic acid (IPB) into the molecules. Followed by 68Ga-, 205/206Bi-, and 177Lu-labelling, the radiopharmaceuticals ([68Ga]Ga-DOTA-IPB-NAPamide, [205/206Bi]Bi-DOTA-IPB-NAPamide, [177Lu]Lu-DOTA-IPB-NAPamide) were characterized in vitro. To test the imaging behavior of the IPB-containing probes, B16F10 tumor-bearing C57BL/6 mice were subjected to in vivo microPET/microSPECT/CT imaging and ex vivo biodistribution studies. All tracers were stable in vitro, with radiochemical purity exceeding 98%. The use of albumin-binding moiety lengthened the in vivo biological half-life of the IPB-carrying radiopharmaceuticals, resulting in elevated tumor accumulation. Both [68Ga]Ga-DOTA-IPB-NAPamide (5.06 ± 1.08 %ID/g) and [205/206Bi]Bi-DOTA-IPB-NAPamide (4.50 ± 0.98 %ID/g) exhibited higher B16F10 tumor concentrations than their matches without the albumin-binding residue ([68Ga]Ga-DOTA-NAPamide and [205/206Bi]Bi-DOTA-NAPamide: 1.18 ± 0.27 %ID/g and 3.14 ± 0.32; respectively), however; the large amounts of off-target radioactivity do not confirm the benefits of half-life extension for short-lived isotopes. Enhanced [177Lu]Lu-DOTA-IPB-NAPamide tumor uptake even 24 h post-injection proved the advantage of IPB-based prolonged circulation time regarding long-lived radionuclides, although the significant background noise must be addressed in this case as well.
Collapse
Affiliation(s)
- Dániel Szücs
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
- Department of Physical Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary;
- Doctoral School of Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Judit P. Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
| | - Viktória Arató
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
- Doctoral School of Pharmaceutical Sciences, Faculty of Pharmacy, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
| | - Barbara Gyuricza
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
| | - Dezső Szikra
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
| | - Imre Tóth
- Department of Physical Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary;
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
| | - Anikó Fekete
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (D.S.); (J.P.S.); (V.A.); (B.G.); (D.S.); (Z.K.); (G.T.)
| |
Collapse
|
30
|
Alati S, Singh R, Pomper MG, Rowe SP, Banerjee SR. Preclinical Development in Radiopharmaceutical Therapy for Prostate Cancer. Semin Nucl Med 2023; 53:663-686. [PMID: 37468417 DOI: 10.1053/j.semnuclmed.2023.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Prostate cancer is a leading cause of cancer death in men worldwide. Among the various treatment options, radiopharmaceutical therapy has shown notable success in metastatic, castration-resistant disease. Radiopharmaceutical therapy is a systemic approach that delivers cytotoxic radiation doses precisely to the malignant tumors and/or tumor microenvironment. Therapeutic radiopharmaceuticals are composed of a therapeutic radionuclide and a high-affinity, tumor-targeting carrier molecule. Therapeutic radionuclides used in preclinical prostate cancer studies are primarily α-, β--, or Auger-electron-emitting radiometals or radiohalogens. Monoclonal antibodies, antibody-derived fragments, peptides, and small molecules are frequently used as tumor-targeting molecules. Over the years, several important membrane-associated proteases and receptors have been identified, validated, and subsequently used for preclinical radiotherapeutic development for prostate cancer. Prostate-specific membrane antigen (PSMA) is the most well-studied prostate cancer-associated protease in preclinical literature. PSMA-targeting radiotherapeutic agents are being investigated using high-affinity antibody- and small-molecule-based agents for safety and efficacy. Early generations of such agents were developed simply by replacing radionuclides of the imaging agents with therapeutic ones. Later, extensive structure-activity relationship studies were conducted to address the safety and efficacy issues obtained from initial patient data. Recent regulatory approval of the 177Lu-labeled low-molecular-weight agent, 177Lu-PSMA-617, is a significant accomplishment. Current preclinical experiments are focused on the structural modification of 177Lu-PSMA-617 and relevant investigational agents to increase tumor targeting and reduce off-target binding and toxicity in healthy organs. While lutetium-177 (177Lu) remains the most widely used radionuclide, radiolabeled analogs with iodine-131 (128I), yttrium-90 (89Y), copper-67 (67Cu), and terbium-161 (161Tb) have been evaluated as potential alternatives in recent years. In addition, agents carrying the α-particle-emitting radiohalogen, astatine-211 (211At), or radiometals, actinium-225 (225Ac), lead-212 (212Pb), radium-223 (223Ra), and thorium-227 (227Th), have been increasingly investigated in preclinical research. Besides PSMA-based radiotherapeutics, other prominent prostate cancer-related proteases, for example, human kallikrein peptidases (HK2 and HK3), have been explored using monoclonal-antibody-(mAb)-based targeting platforms. Several promising mAbs targeting receptors overexpressed on the different stages of prostate cancer have also been developed for radiopharmaceutical therapy, for example, Delta-like ligand 3 (DLL-3), CD46, and CUB domain-containing protein 1 (CDCP1). Progress is also being made using peptide-based targeting platforms for the gastrin-releasing peptide receptor (GRPR), a well-established membrane-associated receptor expressed in localized and metastatic prostate cancers. Furthermore, mechanism-driven combination therapies appear to be a burgeoning area in the context of preclinical prostate cancer radiotherapeutics. Here, we review the current developments related to the preclinical radiopharmaceutical therapy of prostate cancer. These are summarized in two major topics: (1) therapeutic radionuclides and (2) tumor-targeting approaches using monoclonal antibodies, small molecules, and peptides.
Collapse
Affiliation(s)
- Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
31
|
Busslinger SD, Becker AE, Vaccarin C, Deberle LM, Renz ML, Groehn V, Schibli R, Müller C. Investigations Using Albumin Binders to Modify the Tissue Distribution Profile of Radiopharmaceuticals Exemplified with Folate Radioconjugates. Cancers (Basel) 2023; 15:4259. [PMID: 37686538 PMCID: PMC10486429 DOI: 10.3390/cancers15174259] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Introducing an albumin-binding entity into otherwise short-lived radiopharmaceuticals can be an effective means to improve their pharmacokinetic properties due to enhanced blood residence time. In the current study, DOTA-derivatized albumin binders based on 4-(p-iodophenyl)butanoate (DOTA-ALB-1 and DOTA-ALB-3) and 5-(p-iodophenyl)pentanoate entities (DOTA-ALB-24 and DOTA-ALB-25) without and with a hydrophobic 4-(aminomethyl)benzoic acid (AMBA) linker unit, respectively, were synthesized and labeled with lutetium-177 for in vitro and in vivo comparison. Overall, [177Lu]Lu-DOTA-ALB-1 demonstrated ~3-fold stronger in vitro albumin-binding affinity and a longer blood residence time (T50%IA ~8 h) than [177Lu]Lu-DOTA-ALB-24 (T50%IA ~0.8 h). Introducing an AMBA linker enhanced the albumin-binding affinity, resulting in a T50%IA of ~24 h for [177Lu]Lu-DOTA-ALB-3 and ~2 h for [177Lu]Lu-DOTA-ALB-25. The same albumin binders without or with the AMBA linker were incorporated into 6R- and 6S-5-methyltetrahydrofolate-based DOTA-conjugates (177Lu-RedFols). Biodistribution studies in mice performed with both diastereoisomers of [177Lu]Lu-RedFol-1 and [177Lu]Lu-RedFol-3, which comprised the 4-(p-iodophenyl)butanoate moiety, demonstrated a slower accumulation in KB tumors than those of [177Lu]Lu-RedFol-24 and [177Lu]Lu-RedFol-25 with the 5-(p-iodophenyl)pentanoate entity. In all cases, the tumor uptake was high (30-45% IA/g) 24 h after injection. Both diastereoisomers of [177Lu]Lu-RedFol-1 and [177Lu]Lu-RedFol-3 demonstrated high blood retention (3.8-8.7% IA/g, 24 h p.i.) and a 2- to 4-fold lower kidney uptake than the corresponding diastereoisomers of [177Lu]Lu-RedFol-24 and [177Lu]Lu-RedFol-25, which were more rapidly cleared from the blood (<0.2% IA/g, 24 h after injection). Kidney retention of the 6S-diastereoisomers of all 177Lu-RedFols was consistently higher than that of the respective 6R-diastereoisomers, irrespective of the albumin binder and linker unit used. It was demonstrated that the blood clearance data obtained with 177Lu-DOTA-ALBs had predictive value for the blood retention times of the respective folate radioconjugates. The use of these albumin-binding entities without or with an AMBA linker may serve for fine-tuning the blood retention of folate radioconjugates and also other radiopharmaceuticals and, hence, optimize their tissue distribution profiles. Dosimetry estimations based on patient data obtained with one of the most promising folate radioconjugates will be crucial to identify the dose-limiting organ, which will allow for selecting the most suitable folate radioconjugate for therapeutic purposes.
Collapse
Affiliation(s)
- Sarah D. Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Anna E. Becker
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Christian Vaccarin
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Luisa M. Deberle
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
| | - Marie-Luise Renz
- Merck & Cie KmG, Im Laternenacker 5, 8200 Schaffhausen, Switzerland; (M.-L.R.); (V.G.)
| | - Viola Groehn
- Merck & Cie KmG, Im Laternenacker 5, 8200 Schaffhausen, Switzerland; (M.-L.R.); (V.G.)
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland; (S.D.B.); (A.E.B.); (C.V.); (L.M.D.); (R.S.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
32
|
Fu Y, Farnham J, Li W, Powers B, Humphries D, Picard F. LC-MS/MS Bioanalysis of Radioligand Therapeutic Drug Candidate for Preclinical Toxicokinetic Assessment. Anal Chem 2023. [PMID: 37402311 DOI: 10.1021/acs.analchem.3c02163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Radioligand therapy (RLT) has gained significant momentum in recent years in the diagnosis, treatment, and monitoring of cancers. In preclinical development, the safety profile of RLT drug candidate(s) is investigated at relatively low dose levels using the cold (non-radioactive, e.g., 175Lu) ligand as a surrogate of the hot (radioactive, e.g., 177Lu) one in the "ligand-linker-chelator" complex. The formulation of the test article used in preclinical safety studies contains a mixture of free ligand (i.e., ligand-linker-chelator without metal) and cold ligand (i.e., ligand-linker-chelator with non-radioactive metal) in a similar molar ratio as seen under the manufacturing conditions for the RLT drug for clinical use, where only a fraction of free ligand molecules chelate the radioactive metal to form a hot ligand. In this very first report of LC-MS/MS bioanalysis of RLT molecules in support of a regulated preclinical safety assessment study, a highly selective and sensitive LC-MS/MS bioanalytical method was developed for the simultaneous determination of free ligand (NVS001) and cold ligand (175Lu-NVS001) in rat and dog plasma. Several unexpected technical challenges in relation to LC-MS/MS of RLT molecules were successfully addressed. The challenges include poor assay sensitivity of the free ligand NVS001, formation of the free ligand (NVS001) with endogenous metal (e.g., potassium), Ga loss from the Ga-chelated internal standard during sample extraction and analysis, "instability" of the analytes at low concentrations, and inconsistent IS response in the extracted plasma samples. The methods were validated according to the current regulatory requirements in a dynamic range of 0.5-250 ng/mL for both the free and cold ligands using a 25 μL sample volume. The validated method was successfully implemented in sample analysis in support of regulated safety studies, with very good results from incurred sample reanalysis. The current LC-MS/MS workflow can be expanded to quantitative analysis of other RLTs in support of preclinical RLT drug development.
Collapse
Affiliation(s)
- Yunlin Fu
- Pharmacokinetic Sciences─Drug Disposition, Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - James Farnham
- Labcorp Drug Development, 3301 Kinsman Boulevard, Madison, Wisconsin 53704, United States
| | - Wenkui Li
- Pharmacokinetic Sciences─Drug Disposition, Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Brendan Powers
- Labcorp Drug Development, 3301 Kinsman Boulevard, Madison, Wisconsin 53704, United States
| | - David Humphries
- Labcorp Drug Development, 3301 Kinsman Boulevard, Madison, Wisconsin 53704, United States
| | - Franck Picard
- Pharmacokinetic Sciences─Drug Disposition, Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| |
Collapse
|
33
|
Wen X, Xu P, Zeng X, Liu J, Du C, Zeng X, Cheng X, Wang X, Liang Y, Zhao T, Yang H, Li H, Meng L, Fang J, Liu H, Zhou Z, Zhang J, Zhang X, Guo Z, Chen X. Development of [ 177Lu]Lu-LNC1003 for radioligand therapy of prostate cancer with a moderate level of PSMA expression. Eur J Nucl Med Mol Imaging 2023; 50:2846-2860. [PMID: 37097443 DOI: 10.1007/s00259-023-06229-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 04/07/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE Evans blue as an albumin binder has been widely used to improve pharmacokinetics and enhance tumor uptake of radioligands, including prostate-specific membrane antigen (PSMA) targeting agents. The goal of this study is to develop an optimal Evans blue-modified radiotherapeutic agent that could maximize the absolute tumor uptake and tumor absorbed dose thus the therapeutic efficacy to allow treatment of tumors even with moderate level of PSMA expression. METHODS [177Lu]Lu-LNC1003 was synthesized based on PSMA-targeting agent and Evans blue. Binding affinity and PSMA targeting specificity were verified through cell uptake and competition binding assay in 22Rv1 tumor model that has moderate level of PSMA expression. SPECT/CT imaging and biodistribution studies in 22Rv1 tumor-bearing mice were performed to evaluate the preclinical pharmacokinetics. Radioligand therapy studies were conducted to systematically assess the therapeutic effect of [177Lu]Lu-LNC1003. RESULTS LNC1003 showed high binding affinity (IC50 = 10.77 nM) to PSMA in vitro, which was comparable with that of PSMA-617 (IC50 = 27.49 nM) and EB-PSMA-617 (IC50 = 7.91 nM). SPECT imaging of [177Lu]Lu-LNC1003 demonstrated significantly improved tumor uptake and retention as compared with [177Lu]Lu-EB-PSMA and [177Lu]Lu-PSMA-617, making it suitable for prostate cancer therapy. Biodistribution studies further confirmed the remarkably higher tumor uptake of [177Lu]Lu-LNC1003 (138.87 ± 26.53%ID/g) over [177Lu]Lu-EB-PSMA-617 (29.89 ± 8.86%ID/g) and [177Lu]Lu-PSMA-617 (4.28 ± 0.25%ID/g) at 24 h post-injection. Targeted radioligand therapy results showed noteworthy inhibition of 22Rv1 tumor growth after administration of a single dose of 18.5 MBq [177Lu]Lu-LNC1003. There was no obvious antitumor effect after [177Lu]Lu-PSMA-617 treatment under the same condition. CONCLUSION In this study, [177Lu]Lu-LNC1003 was successfully synthesized with high radiochemical purity and stability. High binding affinity and PSMA targeting specificity were identified in vitro and in vivo. With greatly enhanced tumor uptake and retention, [177Lu]Lu-LNC1003 has the potential to improve therapeutic efficacy using significantly lower dosages and less cycles of 177Lu that promises clinical translation to treat prostate cancer with various levels of PSMA expression.
Collapse
Affiliation(s)
- Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Pengfei Xu
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, 272000, China
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Jia Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Chao Du
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Xueyuan Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Xingxing Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Xueqi Wang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Yuanyuan Liang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Hongzhang Yang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Huifeng Li
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Lingxin Meng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Jianyang Fang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Hongwu Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Zijian Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China.
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of PublicHealth, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China.
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| |
Collapse
|
34
|
Lu Q, Long Y, Gai Y, Liu Q, Jiang D, Lan X. [ 177Lu]Lu-PSMA-617 theranostic probe for hepatocellular carcinoma imaging and therapy. Eur J Nucl Med Mol Imaging 2023; 50:2342-2352. [PMID: 36877233 DOI: 10.1007/s00259-023-06155-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/13/2023] [Indexed: 03/07/2023]
Abstract
PURPOSE This study aimed to explore the feasibility of using [177Lu]Lu-prostate-specific membrane antigen (PSMA)-617 and [177Lu]Lu-Evans blue (EB)-PSMA-617 for in vivo radioligand therapy by single-dose administration in a PSMA-positive hepatocellular carcinoma (HCC) xenograft mouse model. METHODS [177Lu]Lu-PSMA-617 and [177Lu]Lu-EB-PSMA-617 were prepared, and labelling efficiency and radiochemical purity were determined. A HepG2 human HCC subcutaneous xenograft mouse model was established. After intravenous injection of [177Lu]Lu-PSMA-617 or [177Lu]Lu-EB-PSMA-617 (37 MBq) into the mouse model, single-photon emission computed tomography/computed tomography (SPECT/CT) was performed. Biodistribution studies were conducted to verify targeting specificity and pharmacokinetics. In the radioligand therapy study, mice were randomized into 4 groups: 37 MBq [177Lu]Lu-PSMA-617, 18.5 MBq [177Lu]Lu-PSMA-617, 7.4 MBq [177Lu]Lu-EB-PSMA-617, and saline (control). A single-dose administration was applied at the beginning of therapy studies. Tumor volume, body weight, and survival were monitored every 2 days. After the end of therapy, mice were euthanized. Tumors were then weighed, and systemic toxicity was evaluated via blood testing and histological examination of healthy organs. RESULTS [177Lu]Lu-PSMA-617 and [177Lu]Lu-EB-PSMA-617 were successfully prepared with high purity and stability. SPECT/CT and biodistribution showed that tumor uptake was higher and persisted longer for [177Lu]Lu-EB-PSMA-617 compared with [177Lu]Lu-PSMA-617. [177Lu]Lu-PSMA-617 was rapidly cleared from the blood, while [177Lu]Lu-EB-PSMA-617 persisted for significantly longer. In radioligand therapy studies, tumor growth was significantly suppressed in the 37 MBq [177Lu]Lu-PSMA-617, 18.5 MBq [177Lu]Lu-PSMA-617, and 7.4 MBq [177Lu]Lu-EB-PSMA-617 groups compared to the saline group. Median survival was 40, 44, 43, and 30 days, respectively. No healthy organ toxicity was observed in safety and tolerability evaluation. CONCLUSIONS Radioligand therapy using [177Lu]Lu-PSMA-617 and [177Lu]Lu-EB-PSMA-617 significantly suppressed tumor growth and prolonged survival time in PSMA-positive HCC xenograft mice without obvious toxicity. These radioligands appear promising for clinical use in humans, and future studies are warranted.
Collapse
Affiliation(s)
- Qiaomiao Lu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, 430022, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China.
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
35
|
Tsuchihashi S, Nakashima K, Tarumizu Y, Ichikawa H, Jinda H, Watanabe H, Ono M. Development of Novel 111In/ 225Ac-Labeled Agent Targeting PSMA for Highly Efficient Cancer Radiotheranostics. J Med Chem 2023. [PMID: 37285471 DOI: 10.1021/acs.jmedchem.3c00346] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Prostate-specific membrane antigen (PSMA) is a promising target for metastatic castration-resistant prostate cancer. We previously reported the effectiveness of PSMA-DA1 as a PSMA-targeting radiotheranostic agent containing an albumin binder moiety. To further enhance tumor uptake, we newly designed PSMA-NAT-DA1 (PNT-DA1) by the introduction of a lipophilic linker into PSMA-DA1. The PSMA affinity of [111In]In-PNT-DA1 was increased (Kd = 8.20 nM) compared with that of [111In]In-PSMA-DA1 (Kd = 89.4 nM). [111In]In-PNT-DA1 showed markedly high tumor accumulation (131.6% injected dose/g at 48 h post-injection), and [111In]In-PNT-DA1 enabled the visualization of the tumor clearly at 24 h post-injection with SPECT/CT imaging. The administration of [225Ac]Ac-PNT-DA1 (2.5 kBq) led to shrinkage of the tumor without marked toxicity, and the antitumor effects of [225Ac]Ac-PNT-DA1 were superior to those of [225Ac]Ac-PSMA-DA1 and [225Ac]Ac-PSMA-617, which is the current gold standard for PSMA-targeting 225Ac-endoradiotherapy. These results suggest that the combination of [111In]In-PNT-DA1 and [225Ac]Ac-PNT-DA1 comprises a promising method of PSMA-targeting radiotheranostics.
Collapse
Affiliation(s)
- Shohei Tsuchihashi
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuta Tarumizu
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Sodegaura-shi, Chiba 299-0266, Japan
| | - Hiroaki Ichikawa
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Sodegaura-shi, Chiba 299-0266, Japan
| | - Hiroki Jinda
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
36
|
Murce E, de Blois E, van den Berg S, de Jong M, Seimbille Y. Synthesis and radiolabelling of PSMA-targeted derivatives containing GYK/MVK cleavable linkers. ROYAL SOCIETY OPEN SCIENCE 2023; 10:220950. [PMID: 36908985 PMCID: PMC9993039 DOI: 10.1098/rsos.220950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Targeted radionuclide therapy (TRT) is a promising strategy to treat different types of cancer. TRT relies on a targeting vector used to deliver a therapeutic radionuclide specifically to the tumour site. Several low molecular weight ligands targeting the prostate-specific membrane antigen (PSMA) have been synthesized, but their pharmacokinetic properties still need to be optimized. Hereby, we describe the synthesis of new conjugates, featuring the cleavable linkers Gly-Tyr-Lys (GYK) and Met-Val-Lys (MVK), to reduce the dose delivered to the kidneys. Compounds were synthesized by solid-phase peptide synthesis (SPPS) and obtained in greater than 95% chemical purity. Radiolabelling was performed with both In-111 and Lu-177 to validate potential use of the compounds as both imaging and therapeutic agents. Radiochemical purity greater than 80% was obtained for both nuclides, but significant radiolysis was observed for the methionine-containing analogue. The results obtained thus far with the GYK-PSMA conjugate could warrant further biological investigations.
Collapse
Affiliation(s)
- Erika Murce
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sophie van den Berg
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- TRIUMF, Life Sciences Division, Vancouver, Canada
| |
Collapse
|
37
|
Wallimann RH, Schindler P, Hensinger H, Tschan VJ, Busslinger SD, Kneuer R, Müller C, Schibli R. Inductively Coupled Plasma Mass Spectrometry─A Valid Method for the Characterization of Metal Conjugates in View of the Development of Radiopharmaceuticals. Mol Pharm 2023; 20:2150-2158. [PMID: 36826437 DOI: 10.1021/acs.molpharmaceut.2c01092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
This study addresses the question whether inductively coupled plasma mass spectrometry (ICP-MS) can be used as a method for the in vitro and in vivo characterization of non-radioactive metal conjugates to predict the properties of analogous radiopharmaceuticals. In a "proof-of-concept" study, the prostate-specific membrane antigen (PSMA)-targeting [175Lu]Lu-PSMA-617 and [159Tb]Tb-PSMA-617 were compared with their respective radiolabeled analogues, [177Lu]Lu-PSMA-617 (PLUVICTO, Novartis) and [161Tb]Tb-PSMA-617. ICP-MS and conventional γ-counting of the cell samples revealed almost identical results (<6% absolute difference between the two technologies) for the in vitro uptake and internalization of the (radio)metal conjugates, irrespective of the employed methodology. In vivo, an equal uptake in PSMA-positive PC-3 PIP tumor xenografts was determined 1 h after the injection of [175Lu]Lu-/[177Lu]Lu-PSMA-617 (41 ± 6% ID/g and 44 ± 12% IA/g, respectively) and [159Tb]Tb-/[161Tb]Tb-PSMA-617 (44 ± 5% ID/g and 44 ± 5% IA/g, respectively). It was further revealed that it is crucial to use the same ratios of the (radio)metal-labeled and unlabeled ligands for both methodologies to obtain equal data in organs in which receptor saturation was reached such as the kidneys (12 ± 2% ID/g vs 10 ± 1% IA/g, 1 h after injection). The data of this study demonstrate that the use of high-sensitivity ICP-MS allows reliable and predictive quantification of compounds labeled with stable metal isotopes in cell and tissue samples obtained in preclinical studies. It can, hence, be employed as a valid alternative to the state-of-the-art γ-counting methodology to detect radioactive ligands.
Collapse
Affiliation(s)
- Rahel H Wallimann
- Novartis Institutes for Biomedical Research, Novartis, 4056 Basel, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Patrick Schindler
- Novartis Institutes for Biomedical Research, Novartis, 4056 Basel, Switzerland
| | - Heloïse Hensinger
- Novartis Institutes for Biomedical Research, Novartis, 4056 Basel, Switzerland
| | - Viviane J Tschan
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Rainer Kneuer
- Novartis Institutes for Biomedical Research, Novartis, 4056 Basel, Switzerland
| | - Cristina Müller
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.,Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Roger Schibli
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.,Center for Radiopharmaceutical Sciences, ETH-PSI, Paul Scherrer Institute, 5232 Villigen, Switzerland
| |
Collapse
|
38
|
Nemtsova ER, Pankratov AA, Morozova NB, Tischenko VK, Petriev VM, Krylov VV, Shegay PV, Ivanov SA, Kaprin AD. Radioligand Therapy of Patients with Metastatic Castrate-Resistant Prostate Cancer. BIOL BULL+ 2022. [DOI: 10.1134/s1062359022120160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
39
|
van der Gaag S, Bartelink IH, Vis AN, Burchell GL, Oprea-Lager DE, Hendrikse H. Pharmacological Optimization of PSMA-Based Radioligand Therapy. Biomedicines 2022; 10:3020. [PMID: 36551776 PMCID: PMC9775864 DOI: 10.3390/biomedicines10123020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy in men of middle and older age. The standard treatment strategy for PCa ranges from active surveillance in low-grade, localized PCa to radical prostatectomy, external beam radiation therapy, hormonal treatment and chemotherapy. Recently, the use of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) for metastatic castration-resistant PCa has been approved. PSMA is predominantly, but not exclusively, expressed on PCa cells. Because of its high expression in PCa, PSMA is a promising target for diagnostics and therapy. To understand the currently used RLT, knowledge about pharmacokinetics (PK) and pharmacodynamics (PD) of the PSMA ligand and the PSMA protein itself is crucial. PK and PD properties of the ligand and its target determine the duration and extent of the effect. Knowledge on the concentration-time profile, the target affinity and target abundance may help to predict the effect of RLT. Increased specific binding of radioligands to PSMA on PCa cells may be associated with better treatment response, where nonspecific binding may increase the risk of toxicity in healthy organs. Optimization of the radioligand, as well as synergistic effects of concomitant agents and an improved dosing strategy, may lead to more individualized treatment and better overall survival.
Collapse
Affiliation(s)
- Suzanne van der Gaag
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Imke H. Bartelink
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Prostate Cancer Network Amsterdam, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - George L. Burchell
- Medical Library, VU University, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Daniela E. Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
40
|
Busslinger SD, Tschan VJ, Richard OK, Talip Z, Schibli R, Müller C. [ 225Ac]Ac-SibuDAB for Targeted Alpha Therapy of Prostate Cancer: Preclinical Evaluation and Comparison with [ 225Ac]Ac-PSMA-617. Cancers (Basel) 2022; 14:5651. [PMID: 36428743 PMCID: PMC9688344 DOI: 10.3390/cancers14225651] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
In the present study, SibuDAB, an albumin-binding PSMA ligand, was investigated in combination with actinium-225 and the data were compared with those of [225Ac]Ac-PSMA-617. In vitro, [225Ac]Ac-SibuDAB and [225Ac]Ac-PSMA-617 showed similar tumor cell uptake and PSMA-binding affinities as their 177Lu-labeled counterparts. The in vitro binding to serum albumin in mouse and human blood plasma, respectively, was 2.8-fold and 1.4-fold increased for [225Ac]Ac-SibuDAB as compared to [177Lu]Lu-SibuDAB. In vivo, this characteristic was reflected by the longer retention of [225Ac]Ac-SibuDAB in the blood than previously seen for [177Lu]Lu-SibuDAB. Similar to [225Ac]Ac-PSMA-617, [225Ac]Ac-SibuDAB was well tolerated at 30 kBq per mouse. Differences in blood cell counts were observed between treated mice and untreated controls, but no major variations were observed between values obtained for [225Ac]Ac-SibuDAB and [225Ac]Ac-PSMA-617. [225Ac]Ac-SibuDAB was considerably more effective to treat PSMA-positive tumor xenografts than [225Ac]Ac-PSMA-617. Only 5 kBq per mouse were sufficient to eradicate the tumors, whereas tumor regrowth was observed for mice treated with 5 kBq [225Ac]Ac-PSMA-617 and only one out of six mice survived until the end of the study. The enhanced therapeutic efficacy of [225Ac]Ac-SibuDAB as compared to that of [225Ac]Ac-PSMA-617 and reasonable safety data qualify this novel radioligand as a candidate for targeted α-therapy of prostate cancer.
Collapse
Affiliation(s)
- Sarah D. Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Viviane J. Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | | | - Zeynep Talip
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 1-5/10, ETH Zurich, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 1-5/10, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
41
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
42
|
Albumin-Mediated Size Exclusion Chromatography: The Apparent Molecular Weight of PSMA Radioligands as Novel Parameter to Estimate Their Blood Clearance Kinetics. Pharmaceuticals (Basel) 2022; 15:ph15091161. [PMID: 36145382 PMCID: PMC9500755 DOI: 10.3390/ph15091161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
A meticulously adjusted pharmacokinetic profile and especially fine-tuned blood clearance kinetics are key characteristics of therapeutic radiopharmaceuticals. We, therefore, aimed to develop a method that allowed the estimation of blood clearance kinetics in vitro. For this purpose, 177Lu-labeled PSMA radioligands were subjected to a SEC column with human serum albumin (HSA) dissolved in a mobile phase. The HSA-mediated retention time of each PSMA ligand generated by this novel 'albumin-mediated size exclusion chromatography' (AMSEC) was converted to a ligand-specific apparent molecular weight (MWapp), and a normalization accounting for unspecific interactions between individual radioligands and the SEC column matrix was applied. The resulting normalized MWapp,norm. could serve to estimate the blood clearance of renally excreted radioligands by means of their influence on the highly size-selective process of glomerular filtration (GF). Based on the correlation between MW and the glomerular sieving coefficients (GSCs) of a set of plasma proteins, GSCcalc values were calculated to assess the relative differences in the expected GF/blood clearance kinetics in vivo and to select lead candidates among the evaluated radioligands. Significant differences in the MWapp,norm. and GSCcalc values, even for stereoisomers, were found, indicating that AMSEC might be a valuable and high-resolution tool for the preclinical selection of therapeutic lead compounds for clinical translation.
Collapse
|
43
|
Koustoulidou S, Handula M, de Ridder C, Stuurman D, Beekman S, de Jong M, Nonnekens J, Seimbille Y. Synthesis and Evaluation of Two Long-Acting SSTR2 Antagonists for Radionuclide Therapy of Neuroendocrine Tumors. Pharmaceuticals (Basel) 2022; 15:ph15091155. [PMID: 36145375 PMCID: PMC9503898 DOI: 10.3390/ph15091155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
Somatostatin receptor subtype 2 (SSTR2) has become an essential target for radionuclide therapy of neuroendocrine tumors (NETs). JR11 was introduced as a promising antagonist peptide to target SSTR2. However, due to its rapid blood clearance, a better pharmacokinetic profile is necessary for more effective treatment. Therefore, two JR11 analogs (8a and 8b), each carrying an albumin binding domain, were designed to prolong the blood residence time of JR11. Both compounds were labeled with lutetium-177 and evaluated via in vitro assays, followed by in vivo SPECT/CT imaging and ex vivo biodistribution studies. [177Lu]Lu-8a and [177Lu]Lu-8b were obtained with high radiochemical purity (>97%) and demonstrated excellent stability in PBS and mouse serum (>95%). [177Lu]Lu-8a showed better affinity towards human albumin compared to [177Lu]Lu-8b. Further, 8a and 8b exhibited binding affinities 30- and 48-fold lower, respectively, than that of the parent peptide JR11, along with high cell uptake and low internalization rate. SPECT/CT imaging verified high tumor accumulation for [177Lu]Lu-8a and [177Lu]Lu-JR11 at 4, 24, 48, and 72 h post-injection, but no tumor uptake was observed for [177Lu]Lu-8b. Ex vivo biodistribution studies revealed high and increasing tumor uptake for [177Lu]Lu-8a. However, its extended blood circulation led to an unfavorable biodistribution profile for radionuclide therapy.
Collapse
Affiliation(s)
- Sofia Koustoulidou
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Savanne Beekman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Julie Nonnekens
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
- Correspondence: ; Tel.: +31-10-703-8961
| |
Collapse
|
44
|
Zha Z, Choi SR, Li L, Zhao R, Ploessl K, Yao X, Alexoff D, Zhu L, Kung HF. New PSMA-Targeting Ligands: Transformation from Diagnosis (Ga-68) to Radionuclide Therapy (Lu-177). J Med Chem 2022; 65:13001-13012. [DOI: 10.1021/acs.jmedchem.2c00852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhihao Zha
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Seok Rye Choi
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Linlin Li
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Ruiyue Zhao
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Karl Ploessl
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Xinyue Yao
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - David Alexoff
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Hank F. Kung
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
45
|
Rosar F, Hügle MJ, Ries M, Bartholomä M, Maus S, Fries P, Khreish F, Ezziddin S. Benefit of including CT urography in [68Ga]PSMA-11 PET/CT with low-dose CT: first results from a larger prostate cancer cohort analysis. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 66:280-289. [PMID: 31992688 DOI: 10.23736/s1824-4785.20.03224-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND Accuracy of [68Ga]PSMA-11 PET/CT may be hampered by ureter accumulation, mimicking lymph node metastases depending on localization and configuration. The benefit of CT urography for differentiation of lymph node metastasis from urinary tract activity was evaluated in a "PET/CT with low-dose CT" setting. METHODS Retrospective analysis of PET/CT for primary staging, biochemical recurrence or local treatment planning in patients with prostate cancer. For CT urography (CTU), iodinated contrast agent was administered 10 minutes prior to image acquisition. All potential pathologic (peri)ureteral tracer uptake was assigned to excretory ureteral accumulation or pathological lesion. To assess additional provided benefit of CTU all foci were rated with an introduced scoring system (ranging from 0 pts: CTU not needed; up to 3 pts: no differentiation possible without CTU). Success of ureter contrasting was assessed by measurement of Hounsfield units. Besides benefit for reading urography-enhanced PET/CT, the possible impact on subsequent patient treatment was evaluated. RESULTS A number of N.=247 patients were included in this study. By CT urography, it was possible to identify each ureter on low-dose CT, with its major part contrasted. In 120/247 (48.6%) patients, urography increased the diagnostic confidence while providing substantial support for interpretation in 60 (24.3%) cases. In 42 (17.0%) patients, urography was clinically relevant (up-/downstaging) with potential impact on subsequent patient care. In 30 of these 42 cases (12.1% of all), discrepant treatment would have resulted from a misdiagnosed tracer accumulation without urography. CONCLUSIONS CT urography benefits the interpretation of [68Ga]-PSMA-11 PET/CT with low-dose CT and leads to discrepant patient treatment in a small but significant subset of patients (12% in our cohort). The implementation of CT urography into standard protocols of [68Ga]PSMA-11 PET/CT with low-dose CT is recommended.
Collapse
Affiliation(s)
- Florian Rosar
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany -
| | - Martin J Hügle
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| | - Martin Ries
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| | - Mark Bartholomä
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| | - Peter Fries
- Clinic of Diagnostic and Interventional Radiology, Saarland University Medical Center, Homburg, Germany
| | - Fadi Khreish
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
46
|
Ding J, Xu M, Chen J, Zhang P, Huo L, Kong Z, Liu Z. 86Y-Labeled Albumin-Binding Fibroblast Activation Protein Inhibitor for Late-Time-Point Cancer Diagnosis. Mol Pharm 2022; 19:3429-3438. [PMID: 35976352 DOI: 10.1021/acs.molpharmaceut.2c00579] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fibroblast activation protein inhibitor (FAPI) is a novel quinoline-based radiopharmaceutical that has theranostic potential, yet the limited tumor retention hinders late-time diagnosis and radionuclide treatment. This study synthesized four albumin-binding FAPIs (TE-FAPI-01 to 04) and evaluated their in vitro stability, binding affinity, in vivo biodistribution, and tumor uptake with 68Ga, 86Y, and 177Lu labeling, aiming to select the best molecule that has favorable pharmacokinetics to extend the blood circulation and tumor uptake in FAP-expressing tumors. All TE-FAPIs were stable in saline and plasma and displayed high FAP-binding affinity, with IC50 values ranging from 3.96 to 34.9 nmol/L. The capabilities of TE-FAPIs to be retained in circulation were higher than that of FAPI-04, and TE-FAPI-04 displayed minimum physiological uptake in major organs compared with other molecules. TE-FAPI-03 and TE-FAPI-04 exhibited persistent tumor accumulation, with tumor radioactivity 24 h after administration of 2.84 ± 1.19%ID/g and 3.86 ± 1.15%ID/g for 177Lu-TE-FAPI-03 and 177Lu-TE-FAPI-04, respectively, both of which outperformed 177Lu-FAPI-04 (0.34 ± 0.07%ID/g). TE-FAPI-04 was recognized as the albumin-binding FAPI with the most favorable pharmacokinetics and imaging performance. The enhanced circulation half-life and tumor uptake of TE-FAPI-04 aided the theranostics of malignant tumors and warrant further clinical investigations.
Collapse
Affiliation(s)
- Jie Ding
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Mengxin Xu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Junyi Chen
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Pu Zhang
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ziren Kong
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhibo Liu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Peking University-Tsinghua University Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
47
|
Hu B, Liu T, Li L, Shi L, Yao M, Li C, Ma X, Zhu H, Jia B, Wang F. IgG-Binding Nanobody Capable of Prolonging Nanobody-Based Radiotracer Plasma Half-Life and Enhancing the Efficacy of Tumor-Targeted Radionuclide Therapy. Bioconjug Chem 2022; 33:1328-1339. [PMID: 35687724 DOI: 10.1021/acs.bioconjchem.2c00209] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nanobodies have been developed rapidly as targeted probes for molecular imaging owing to their high affinity, outstanding tissue penetration, and rapid blood clearance. However, the short retention time at the tumor site limits their application in targeted radionuclide therapy. In this study, we designed a dual-targeting nanobody referred to as MIRC213-709, which can specifically bind to the HER2 receptor in tumor cell lines with high affinity (by nanobody MIRC213) and endogenous IgG in plasma to prolong the half-life by the MIRC213 C-terminal fusion nanobody, MIRC709. The nanobodies were site-specifically radiolabeled with 99mTc and 177Lu, and radiochemical purity was >95% after purification. The long blood circulation time and tumor retention property of 99mTc/177Lu-MIRC213-709 were confirmed by a blood clearance assay, single-photon emission computed tomography (SPECT), and a biodistribution study. The blood clearance assay showed that the distribution phase half-life (T1/2α) and elimination phase half-life (T1/2β) of 99mTc-MIRC213-709 were 6.74- and 19.04-fold longer than those of 99mTc-MIRC213, respectively. The SPECT/CT and biodistribution results showed that the highest uptake of 177Lu-MIRC213 in the NCI-N87 model was 5.24 ± 0.95% ID/g at 6 h p.i., while the highest uptake of 177Lu-MIRC213-709 in the NCI-N87 model was 30.82 ± 7.29% ID/g at 48 h p.i. Compared with 177Lu-MIRC213, 177Lu-MIRC213-709 had a 16.9-fold increased tumor cumulative uptake (2606 ± 195.1 vs 153.9 ± 22.37% ID/g·h). The targeted radionuclide therapy assay was performed in the NCI-N87 tumor model, and treatment monitoring ended on day 32. The post-treatment/pretreatment tumor volumes were 12.99 ± 1.66, 3.58 ± 0.96, 1.26 ± 0.17, and 1.54 ± 0.50 in the 0, 9, and 18 MBq single-dose groups and the two 9 MBq divided dose group (14 days apart), respectively. All treatment groups showed significant therapeutic effects (P < 0.0001). Thus, fusion with the IgG-binding nanobody MIRC709 provides MIRC213 derivatives with improved metabolic properties for targeted radionuclide therapy.
Collapse
Affiliation(s)
- Biao Hu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Liqiang Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Linqing Shi
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Meinan Yao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chenzhen Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
48
|
Iikuni S, Tarumizu Y, Tsuchihashi S, Ohara T, Watanabe H, Ono M. Synthesis and Evaluation of Novel 111In-Labeled Picolinic Acid-Based Radioligands Containing an Albumin Binder for Development of a Radiotheranostic Platform. Mol Pharm 2022; 19:2725-2736. [PMID: 35758049 DOI: 10.1021/acs.molpharmaceut.2c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Picolinic acid-based metallic chelators, e.g., neunpa and octapa, have attracted much attention as promising scaffolds for radiotheranostic agents, particularly those containing larger α-emitting radiometals. Furthermore, albumin binder (ALB) moieties, which noncovalently bind to albumin, have been utilized to improve the pharmacokinetics of radioligands targeting various biomolecules. In this study, we designed and synthesized novel neunpa and octapa derivatives (Neunpa-2 and Octapa-2, respectively), which contained a prostate-specific membrane antigen (PSMA)-binding moiety (model targeting vector) and an ALB moiety. We evaluated the fundamental properties of these derivatives as radiotheranostic agents using 111In. In a cell-binding assay using LNCaP (PSMA-positive) cells, [111In]In-Neunpa-2 and [111In]In-Octapa-2 specifically bound to the LNCaP cells. In addition, a human serum albumin (HSA)-binding assay revealed that [111In]In-Neunpa-2 and [111In]In-Octapa-2 exhibited greater binding to HSA than their non-ALB-conjugated counterparts ([111In]In-Neunpa-1 and [111In]In-Octapa-1, respectively). A biodistribution assay conducted in LNCaP tumor-bearing mice showed that the introduction of the ALB moiety into the 111In-labeled neunpa and octapa derivatives resulted in markedly enhanced tumor uptake and retention of the radioligands. Furthermore, single-photon emission computed tomography imaging of LNCaP tumor-bearing mice with [111In]In-Octapa-2 produced tumor images. These results indicate that [111In]In-Octapa-2 may be a useful PSMA imaging probe and that picolinic acid-based ALB-conjugated radiometallic complexes may be promising candidates as radiotheranostic agents.
Collapse
Affiliation(s)
- Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501 Kyoto, Japan
| | - Yuta Tarumizu
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501 Kyoto, Japan
| | - Shohei Tsuchihashi
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501 Kyoto, Japan
| | - Takaki Ohara
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501 Kyoto, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501 Kyoto, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501 Kyoto, Japan
| |
Collapse
|
49
|
Tschan VJ, Borgna F, Busslinger SD, Stirn M, Rodriguez JMM, Bernhardt P, Schibli R, Müller C. Preclinical investigations using [177Lu]Lu-Ibu-DAB-PSMA toward its clinical translation for radioligand therapy of prostate cancer. Eur J Nucl Med Mol Imaging 2022; 49:3639-3650. [PMID: 35635566 PMCID: PMC9399046 DOI: 10.1007/s00259-022-05837-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 05/08/2022] [Indexed: 12/21/2022]
Abstract
Abstract[177Lu]Lu-Ibu-DAB-PSMA was previously characterized with moderate albumin-binding properties enabling high tumor accumulation but reasonably low retention in the blood. The aim of this study was to investigate [177Lu]Lu-Ibu-DAB-PSMA in preclinical in vivo experiments and compare its therapeutic efficacy and potential undesired side effects with those of [177Lu]Lu-PSMA-617 and the previously developed [177Lu]Lu-PSMA-ALB-56. BALB/c nude mice without tumors were investigated on Day 10 and 28 after injection of 10 MBq radioligand. It was revealed that most plasma parameters were in the same range for all groups of mice and histopathological examinations of healthy tissue did not show any alternations in treated mice as compared to untreated controls. Based on these results, a therapy study over twelve weeks was conducted with PC-3 PIP tumor-bearing mice for comparison of the radioligands’s therapeutic efficacy up to an activity of 10 MBq (1 nmol) per mouse. In agreement with the increased mean absorbed tumor dose, [177Lu]Lu-Ibu-DAB-PSMA (~ 6.6 Gy/MBq) was more effective to inhibit tumor growth than [177Lu]Lu-PSMA-617 (~ 4.5 Gy/MBq) and only moderately less potent than [177Lu]Lu-PSMA-ALB-56 (~ 8.1 Gy/MBq). As a result, the survival of mice treated with 2 MBq of an albumin-binding radioligand was significantly increased (p < 0.05) compared to that of mice injected with [177Lu]Lu-PSMA-617 or untreated controls. The majority of mice treated with 5 MBq or 10 MBq [177Lu]Lu-Ibu-DAB-PSMA or [177Lu]Lu-PSMA-ALB-56 were still alive at study end. Hemograms of immunocompetent mice injected with 30 MBq [177Lu]Lu-Ibu-DAB-PSMA or 30 MBq [177Lu]Lu-PSMA-617 showed values in the same range as untreated controls. This was, however, not the case for mice treated with [177Lu]Lu-PSMA-ALB-56 which revealed a drop in lymphocytes and hemoglobin at Day 10 and Day 28 after injection. The data of this study demonstrated a significant therapeutic advantage of [177Lu]Lu-Ibu-DAB-PSMA over [177Lu]Lu-PSMA-617 and a more favorable safety profile as compared to that of [177Lu]Lu-PSMA-ALB-56. Based on these results, [177Lu]Lu-Ibu-DAB-PSMA may has the potential for a clinical translation.
Collapse
Affiliation(s)
- Viviane J Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Francesca Borgna
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Martina Stirn
- Clinical Laboratory, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Josep M Monné Rodriguez
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Peter Bernhardt
- Department of Radiation Physics, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland.
| |
Collapse
|
50
|
Jeitner TM, Babich JW, Kelly JM. Advances in PSMA theranostics. Transl Oncol 2022; 22:101450. [PMID: 35597190 PMCID: PMC9123266 DOI: 10.1016/j.tranon.2022.101450] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 12/15/2022] Open
Abstract
PSMA is an appealing target for theranostic because it is a transmembrane protein with a known substrate that is overexpessed on prostate cancer cells and internalizes upon ligand binding. There are a number of PSMA theranostic ligands in clinical evaluation, clinical trial, or clinically approved. PSMA theranostic ligands increase progression-free survival, overall survival, and pain in patients with metastatic castration resistant prostate cancer. A major obstacle to PSMA-targeted radioligand therapy is off-target toxicity in salivary glands.
The validation of prostate specific membrane antigen (PSMA) as a molecular target in metastatic castration-resistant prostate cancer has stimulated the development of multiple classes of theranostic ligands that specifically target PSMA. Theranostic ligands are used to image disease or selectively deliver cytotoxic radioactivity to cells expressing PSMA according to the radioisotope conjugated to the ligand. PSMA theranostics is a rapidly advancing field that is now integrating into clinical management of prostate cancer patients. In this review we summarize published research describing the biological role(s) and activity of PSMA, highlight the most clinically advanced PSMA targeting molecules and biomacromolecules, and identify next generation PSMA ligands that aim to further improve treatment efficacy. The goal of this review is to provide a comprehensive assessment of the current state-of-play and a roadmap to achieving further advances in PSMA theranostics.
Collapse
Affiliation(s)
- Thomas M Jeitner
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA
| | - John W Babich
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA; Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, New York, NY 10021, USA; Weill Cornell Medicine, Citigroup Biomedical Imaging Center, New York, NY 10021, USA
| | - James M Kelly
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA; Weill Cornell Medicine, Citigroup Biomedical Imaging Center, New York, NY 10021, USA.
| |
Collapse
|