1
|
Kazuta N, Nakashima K, Tarumizu Y, Sato T, Maya Y, Watanabe H, Ono M. Novel Radiotheranostic Ligands Targeting Prostate-Specific Membrane Antigen Based on Dual Linker Approach. Mol Pharm 2025; 22:377-386. [PMID: 39614820 DOI: 10.1021/acs.molpharmaceut.4c00974] [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: 01/07/2025]
Abstract
Radiotheranostics using prostate-specific membrane antigen (PSMA)-targeting radioligands offers precision medicine by performing radionuclide therapy based on results of diagnosis. Albumin binder (ALB) binds to albumin reversibly and contributes to effective radiotheranostics by enhancing tumor accumulation of PSMA-targeting radioligands. We newly developed two ALB-containing PSMA-targeting radioligands including dual functional linkers, a hydrophilic linker, d-glutamic acid, and a hydrophobic linker, 4-(aminomethyl)benzoic acid, with the opposite arrangement (PNT-DA6 and PNT-DA7). A biodistribution study of [111In]In-PNT-DA6 indicated that the introduction and arrangement of dual functional linkers contributed to improved pharmacokinetics. A single photon emission computed tomography study of [111In]In-PNT-DA6 produced a clear PSMA-expressing tumor image. Moreover, [225Ac]Ac-PNT-DA6 showed the inhibition of tumor growth in targeted radionuclide therapy in PSMA-expressing tumor-bearing mice. These results indicated that [111In]In-PNT-DA6 and [225Ac]Ac-PNT-DA6 exhibited useful characteristics as PSMA-targeting radiotheranostic ligands.
Collapse
Affiliation(s)
- Nobuki Kazuta
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuta Tarumizu
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Kitasode, Sodegaura-shi, Chiba 299-0266, Japan
| | - Takumi Sato
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Kitasode, Sodegaura-shi, Chiba 299-0266, Japan
| | - Yoshifumi Maya
- Research Center, Nihon Medi-Physics Co., Ltd., 3-1 Kitasode, Sodegaura-shi, Chiba 299-0266, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
2
|
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
|
3
|
Chen J, Pang Y, Liao X, Zhou Y, Luo Q, Wu H, Zuo C, Zhang J, Lin Q, Chen X, Zhao L, Chen H. Development of [ 177Lu]Lu-LNC1010 for peptide receptor radionuclide therapy of nasopharyngeal carcinoma. Eur J Nucl Med Mol Imaging 2024; 52:247-259. [PMID: 39145784 DOI: 10.1007/s00259-024-06874-9] [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: 05/06/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE Somatostatin Receptor 2 (SSTR2)-targeted radiopharmaceutical [68Ga]Ga-DOTATATE has potential advantages in the diagnosis of nasopharyngeal carcinoma (NPC). This study introduces a novel long-lasting SSTR2 analogue, LNC1010, based on DOTATATE, a truncated Evans blue-binding moiety, and a polyethylene-glycol linker. We hypothesised that peptide receptor radionuclide therapy (PRRT) is more effective with [177Lu]Lu-LNC1010 than with [177Lu]Lu-DOTATATE in treating metastatic NPC. METHODS We assessed binding characteristics of LNC1010 in vitro using C666-1 NPC cells and in-vivo pharmacokinetics of [68Ga]Ga/[177Lu]Lu-LNC1010 in C666-1 NPC xenografts via PET and SPECT imaging, biodistribution studies, and PRRT, and compared them with [68Ga]Ga/[177Lu] Lu-labelled DOTATATE. Furthermore, a proof-of-concept approach for imaging and therapy was conducted in a patient with metastatic NPC. RESULTS LNC1010 exhibited strong uptake and specific affinity for SSTR2 in C666-1 NPC cells. PET and SPECT imaging demonstrated higher uptake and longer tumour retention of [68Ga]Ga/[177Lu]Lu-LNC1010 than [68Ga]Ga/[177Lu]Lu-DOTATATE in C666-1 NPC xenografts, indicating its suitability for PRRT applications in NPCs. Biodistribution studies confirmed the higher uptake and prolonged retention of [177Lu]Lu-LNC1010 than [177Lu]Lu-DOTATATE. In preclinical PRRT studies, [177Lu]Lu-LNC1010 showed greater inhibition of tumour growth in C666-1 NPC xenografts than [177Lu]Lu-DOTATATE. In a subsequent pilot clinical study, PRRT with [177Lu]Lu-LNC1010 achieved favourable therapeutic and negligible side effects in a patient with metastatic NPC. CONCLUSION [177Lu]Lu-LNC1010 demonstrated increased tumour uptake and prolonged retention in SSTR2-positive NPCs, with superior anti-tumour efficacy to that of [177Lu]Lu-DOTATATE in preclinical studies. These findings suggest that PRRT with [177Lu]Lu-LNC1010 is a promising treatment for advanced NPC, extending the clinical scope of PRRT beyond neuroendocrine tumours.
Collapse
Affiliation(s)
- Jianhao Chen
- Department of Nuclear Medicine and Minnan PET Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yizhen Pang
- Department of Nuclear Medicine and Minnan PET Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Xiyi Liao
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yangfan Zhou
- Department of Nuclear Medicine and Minnan PET Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qicong Luo
- Laboratory of Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hua Wu
- Department of Nuclear Medicine and Minnan PET Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changjing Zuo
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Military Medical University (Shanghai Changhai Hospital), Shanghai, China
| | - Jingjing Zhang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Liang Zhao
- Department of Nuclear Medicine and Minnan PET Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Haojun Chen
- Department of Nuclear Medicine and Minnan PET Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
4
|
Bauer IA, Dmitrienko EV. Amphiphilic Oligonucleotide Derivatives-Promising Tools for Therapeutics. Pharmaceutics 2024; 16:1447. [PMID: 39598570 PMCID: PMC11597563 DOI: 10.3390/pharmaceutics16111447] [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: 10/18/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Recent advances in genetics and nucleic acid chemistry have created fundamentally new tools, both for practical applications in therapy and diagnostics and for fundamental genome editing tasks. Nucleic acid-based therapeutic agents offer a distinct advantage of selectively targeting the underlying cause of the disease. Nevertheless, despite the success achieved thus far, there remain unresolved issues regarding the improvement of the pharmacokinetic properties of therapeutic nucleic acids while preserving their biological activity. In order to address these challenges, there is a growing focus on the study of safe and effective delivery methods utilising modified nucleic acid analogues and their lipid bioconjugates. The present review article provides an overview of the current state of the art in the use of chemically modified nucleic acid derivatives for therapeutic applications, with a particular focus on oligonucleotides conjugated to lipid moieties. A systematic analysis has been conducted to investigate the ability of amphiphilic oligonucleotides to self-assemble into micelle-like structures, as well as the influence of non-covalent interactions of such derivatives with serum albumin on their biodistribution and therapeutic effects.
Collapse
Affiliation(s)
| | - Elena V. Dmitrienko
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
| |
Collapse
|
5
|
Yu Z, Jiang Z, Cheng X, Yuan L, Chen H, Ai L, Wu Z. Development of fibroblast activation protein-α radiopharmaceuticals: Recent advances and perspectives. Eur J Med Chem 2024; 277:116787. [PMID: 39197253 DOI: 10.1016/j.ejmech.2024.116787] [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: 07/23/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Fibroblast activation protein-α (FAP) has emerged as a promising target in the field of radiopharmaceuticals due to its selective expression in cancer-associated fibroblasts (CAFs) and other pathological conditions involving fibrosis and inflammation. Recent advancements have focused on developing FAP-specific radioligands for diagnostic imaging and targeted radionuclide therapy. This perspective summarized the latest progress in FAP radiopharmaceutical development, highlighting novel radioligands, preclinical evaluations, and potential clinical applications. Additionally, we analyzed the advantages and existing problems of targeted FAP radiopharmaceuticals, and discussed the key breakthrough directions of this target, so as to improve the development and conversion of FAP-targeted radiopharmaceuticals.
Collapse
Affiliation(s)
- Ziyue Yu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Zeng Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Xuebo Cheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Leilei Yuan
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Lin Ai
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Zehui Wu
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
6
|
Yamaguchi K, Kazuta N, Tsuchihashi S, Watanabe H, Ono M. Structure-affinity-pharmacokinetics relationships of 111In-labeled PSMA-targeted ligands with different albumin binders. Nucl Med Biol 2024; 138-139:108945. [PMID: 39153354 DOI: 10.1016/j.nucmedbio.2024.108945] [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: 04/20/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) is a promising target for treating metastatic castration-resistant prostate cancer. Our previous report presented 111In- or 225Ac-labeled PSMA-NAT-DA1 (PNT-DA1) as a PSMA-targeted ligand. To improve its therapeutic efficiency, PNT-DA1 contains 4-(p-iodophenyl)butyric acid (IPBA), which is known as an albumin binder (ALB) moiety. However, few reports have examined the relationship between the chemical modification of the ALB moiety and pharmacokinetics of PSMA-targeted radioligands. To assess this relationship, we designed, synthesized, and evaluated four [111In]In-PNT-DA1 analogues with ALB moieties different from IPBA. METHODS The [111In]In-PNT-DA1 analogues were synthesized from their corresponding precursors through ligand substitution reaction. The stability of [111In]In-PNT-DA1 analogues in mouse plasma, their affinity for human serum albumin (HSA), their binding to mouse plasma proteins, and their affinity for PSMA were evaluated in vitro. The tissue distribution profile of the radioligands was assessed in biodistribution studies using LNCaP tumor-bearing nude mice. RESULTS All [111In]In-PNT-DA1 analogues were obtained at a high radiochemical yield and purity. These analogues were highly stable in mouse plasma after 24 h. The binding affinity for HSA significantly varied among the different ALB moieties. Moreover, high affinity for mouse plasma proteins was observed for all [111In]In-PNT-DA1 analogues compared with their counterparts without an ALB moiety. The affinity for PSMA was comparable for all radioligands. In the biodistribution assay, the pharmacokinetics of [111In]In-PNT-DA1 analogues varied markedly depending on the type of ALB moiety. In particular, tumor area under the curve (AUC) values were increased for radioligands with higher blood retention, while some previous studies reported that compounds with moderate blood retention exhibited the highest tumor AUC values. CONCLUSION The introduction of an appropriate ALB moiety into the ligand may lead to the development of more useful PSMA-targeted radioligands with higher tumor accumulation.
Collapse
Affiliation(s)
- Keisei Yamaguchi
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Nobuki Kazuta
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - 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
| | - 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
|
7
|
Mahmoud Shokhba AS, El-Deen A Omran MA, Abdel-Rahman MA, El-Shenawy NS. Effect of Egyptian spitting cobra Naja nubiae crude venom on immunogenic activity of rats. Toxicon 2024; 247:107834. [PMID: 38950737 DOI: 10.1016/j.toxicon.2024.107834] [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: 04/12/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Snakes show defensive activities, often counting visual or auditory displays against an aggressor. The study observed what happens to rats administered subcutaneously sub-lethal doses of crude venom Naja nubiae. The pro-inflammatory cytokines, such as tumor necrosis alpha (TNF-α) and interleukin-6 (IL-6), as well as the anti-inflammatory cytokines such as interleukin-10 (IL-10), and inflammatory mediator's prostaglandin E-2 (PG-E2), were evaluated. Vascular permeability (VP) was employed to assess how leaky or permeable blood vessels are in various tissues and organs, including the rat peritoneal cavity and lymphoid organs. Lymphoid organs' histological alterations brought on by Nubiae venom. The study found that the two venom doses-1/4 and 1/2 LD50-induced high levels of inflammatory activity as evidenced by the production of inflammatory cytokines. These findings demonstrated that venom enhanced innate immunity through specifically increased T helper cells, IL-6, TNF-α, IL-10, and PG-E2. The results reveal whether the venom has an immunomodulatory effect and promotes inflammation. The data have a substantial impact on the development of new drugs and treatments for inflammatory conditions.
Collapse
|
8
|
Kazuta N, Nakashima K, Watanabe H, Ono M. Effect of Linker Entities on Pharmacokinetics of 111In-Labeled Prostate-Specific Membrane Antigen-Targeting Ligands with an Albumin Binder. ACS Pharmacol Transl Sci 2024; 7:2401-2413. [PMID: 39144550 PMCID: PMC11320743 DOI: 10.1021/acsptsci.4c00257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024]
Abstract
In the field of radiopharmaceutical development targeting cancer, an albumin binder (ALB) is commonly used to improve accumulation of radioligands in tumors because it has high binding affinity for albumin and extends the circulation time of radioligands. The further development of ALB-containing radioligands is also expected to regulate their pharmacokinetics. In this study, we newly designed and synthesized [111In]In-PNT-DA1 derivatives, prostate-specific membrane antigen (PSMA)-targeting radioligands including a functional linker (d-glutamic acid or 4-(aminomethyl)benzoic acid), and evaluated the relationships among the structure, albumin-binding affinity, and pharmacokinetics. These derivatives showed a different binding affinity for albumin by the introduction of a linker. Biodistribution studies revealed that the introduction of a linker affects the pharmacokinetics of each derivative. The biodistribution studies also suggested that moderate albumin-binding affinity enhances the tumor/kidney ratio of the derivative. SPECT imaging using [111In]In-PNT-DA3 with the highest tumor/kidney ratio among [111In]In-PNT-DA1 derivatives led to clear visualization of a PSMA-positive LNCaP tumor. The results suggest that the appropriate introduction of linker entities may be necessary to improve the pharmacokinetics of PSMA-targeting radioligands.
Collapse
Affiliation(s)
- Nobuki Kazuta
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| | - Kazuma Nakashima
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Sakyo-ku 606-8501, Japan
| |
Collapse
|
9
|
Echigo H, Munekane M, Fuchigami T, Washiyama K, Mishiro K, Wakabayashi H, Takahashi K, Kinuya S, Ogawa K. Optimizing the pharmacokinetics of an 211At-labeled RGD peptide with an albumin-binding moiety via the administration of an albumin-binding inhibitor. Eur J Nucl Med Mol Imaging 2024; 51:2663-2671. [PMID: 38570359 PMCID: PMC11224111 DOI: 10.1007/s00259-024-06695-w] [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: 01/06/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE A probe for targeted alpha therapy (TAT) using the RGD peptide (Ga-DOTA-K([211At]APBA)-c(RGDfK) ([211At]1)) with albumin-binding moiety (ABM) was recently developed. [211At]1 highly accumulated in tumors and significantly inhibited tumor growth in U-87 MG tumor-bearing mice. However, high [211At]1 retention in blood may cause critical adverse events, such as hematotoxicity. Therefore, we attempted to accelerate the blood clearance of [211At]1 by competitively inhibiting the binding of [211At]1 to albumin to modulate the pharmacokinetics of the former. METHODS To evaluate the effects of albumin-binding inhibitors in normal mice, sodium 4-(4-iodophenyl)butanoate at 2, 5, or 10 molar equivalents of blood albumin was administered at 1-h postinjection of [211At]1. The biodistribution of [211At]1, SPECT/CT imaging of [67Ga]Ga-DOTA-K(IPBA)-c(RGDfK) ([67Ga]2), and the therapeutic effects of [211At]1 were compared with or without IPBA administration in U-87 MG tumor-bearing mice. RESULTS Blood radioactivity of [211At]1 was decreased in a dose-dependent manner with IPBA in normal mice. In U-87 MG tumor-bearing mice, the blood radioactivity and accumulation in nontarget tissues of [211At]1 were decreased by IPBA. Meanwhile, tumor [211At]1 accumulation was not changed at 3-h postinjection of IPBA. In SPECT/CT imaging of [67Ga]2, IPBA administration dramatically decreased radioactivity in nontarget tissues, and only tumor tissue was visualized. In therapeutic experiments, [211At]1 with IPBA injected-group significantly inhibited tumor growth compared to the control group. CONCLUSION IPBA administration (as an albumin-binding inhibitor) could modulate the pharmacokinetics and enhance the therapeutic effects of [211At]1.
Collapse
Affiliation(s)
- Hiroaki Echigo
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Ishikawa, Japan
| | - Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Ishikawa, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Ishikawa, Japan
| | - Kohshin Washiyama
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Ishikawa, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-Machi 13-1, Kanazawa, 920-8641, Ishikawa, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-Machi 13-1, Kanazawa, 920-8641, Ishikawa, Japan
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Ishikawa, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Ishikawa, Japan.
| |
Collapse
|
10
|
Kazuta N, Tsuchihashi S, Watanabe H, Ono M. Fundamental evaluation regarding the relationship between albumin-binding and tumor accumulation of PSMA-targeting radioligands. Ann Nucl Med 2024; 38:574-583. [PMID: 38676906 DOI: 10.1007/s12149-024-01930-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/08/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVE The marked success of prostate-specific membrane antigen (PSMA)-targeting radioligands with albumin binder (ALB) is attributed to the improvement of blood retention and tumor accumulation. [111In]In-PNT-DA1, our PSMA-targeting radioligand with ALB, also achieved improved tumor accumulation due to its prolonged blood retention. Although the advantage of ALBs is related to their reversible binding to albumin, the relationship between albumin-binding and tumor accumulation of PSMA-targeting radioligands remains unclear because of the lack of information about radioligands with stronger albumin-binding than ALBs. In this study, we designed and synthesized [111In]In-PNT-DM-HSA, a new radioligand that consists of a PSMA-targeting radioligand covalently bound to albumin. The pharmacokinetics of [111In]In-PNT-DM-HSA was compared with those of [111In]In-PNT-DA1 and [111In]In-PSMA-617, a non-ALB-conjugated radioligand, to evaluate the relationship between albumin-binding and tumor accumulation. METHOD The [111In]In-PNT-DM-HSA was prepared by incubation of [111In]In-PNT-DM, a PSMA-targeting radioligand including a maleimide group, and human serum albumin (HSA). The ability of [111In]In-PNT-DM-HSA was evaluated by in vitro assays. A biodistribution study using LNCaP tumor-bearing mice was conducted to compare the pharmacokinetics of [111In]In-PNT-DM-HSA, [111In]In-PNT-DA1, and [111In]In-PSMA-617. RESULTS The [111In]In-PNT-DM-HSA was obtained at a favorable radiochemical yield and high radiochemical purity. In vitro assays revealed that [111In]In-PNT-DM-HSA had fundamental characteristics as a PSMA-targeting radioligand interacting with albumin covalently. In a biodistribution study, [111In]In-PNT-DM-HSA and [111In]In-PNT-DA1 showed higher blood retention than [111In]In-PSMA-617. On the other hand, the tumor accumulation of [111In]In-PNT-DA1 was much higher than [111In]In-PNT-DM-HSA and [111In]In-PSMA-617. CONCLUSIONS These results indicate that the moderate reversible binding of ALB with albumin, not covalent binding, may play a critical role in enhancing the tumor accumulation of PSMA-targeting radioligands.
Collapse
Affiliation(s)
- Nobuki Kazuta
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-Cho, Sakyo-Ku, Kyoto, 606-8501, Japan
| | - 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
| | - 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
|
11
|
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
|
12
|
Munekane M, Fuchigami T, Ogawa K. Recent advances in the development of 225Ac- and 211At-labeled radioligands for radiotheranostics. ANAL SCI 2024; 40:803-826. [PMID: 38564087 PMCID: PMC11035452 DOI: 10.1007/s44211-024-00514-w] [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: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 04/04/2024]
Abstract
Radiotheranostics utilizes a set of radioligands incorporating diagnostic or therapeutic radionuclides to achieve both diagnosis and therapy. Imaging probes using diagnostic radionuclides have been used for systemic cancer imaging. Integration of therapeutic radionuclides into the imaging probes serves as potent agents for radionuclide therapy. Among them, targeted alpha therapy (TAT) is a promising next-generation cancer therapy. The α-particles emitted by the radioligands used in TAT result in a high linear energy transfer over a short range, inducing substantial damage to nearby cells surrounding the binding site. Therefore, the key to successful cancer treatment with minimal side effects by TAT depends on the selective delivery of radioligands to their targets. Recently, TAT agents targeting biomolecules highly expressed in various cancer cells, such as sodium/iodide symporter, norepinephrine transporter, somatostatin receptor, αvβ3 integrin, prostate-specific membrane antigen, fibroblast-activation protein, and human epidermal growth factor receptor 2 have been developed and have made remarkable progress toward clinical application. In this review, we focus on two radionuclides, 225Ac and 211At, which are expected to have a wide range of applications in TAT. We also introduce recent fundamental and clinical studies of radiopharmaceuticals labeled with these radionuclides.
Collapse
Affiliation(s)
- Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
13
|
Dai D, Yu J, Gou W, Yang S, Li Y, Wang Z, Yang Z, Huang T, Li P, Zhu T, Hou W, Zhao Y, Xu W, Li Y. Novel CDK19-Targeted Radiotracers: A Potential Design Strategy to Improve the Pharmacokinetics and Tumor Uptake. J Med Chem 2024; 67:6726-6737. [PMID: 38570733 DOI: 10.1021/acs.jmedchem.4c00281] [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: 04/05/2024]
Abstract
Cyclin-dependent kinase 19 (CDK19) is overexpressed in prostate cancer, making it an attractive target for both imaging and therapy. Since little is known about the optimized approach for radioligands of nuclear proteins, linker optimization strategies were used to improve pharmacokinetics and tumor absorption, including the adjustment of the length, flexibility/rigidity, and hydrophilicity/lipophilicity of linkers. Molecular docking was conducted for virtual screening and followed by IC50 determination. Both BALB/c mice and P-16 xenografts were used for tissue distribution and PET/CT imaging. The ligand 68Ga-10c demonstrated high absorption in tumor 5 min after injection and sustains long-term imaging within 3 h. Furthermore, 68Ga-10c exhibited slow clearance within the tumor and was predominantly metabolized in both the liver and kidneys, showing the potential to alleviate metabolic pressure and enhance tissue safety. Therefore, the linker optimization strategy is well suited for CDK19 and provides a reference for the radioactive ligands of other nuclear targets.
Collapse
Affiliation(s)
- 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 300060, China
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, 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 300192, China
| | - Wenfeng Gou
- 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 300192, China
| | - Shuangmeng Yang
- 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 300192, 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 300192, China
| | - Ziyang Wang
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, 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 300060, China
| | - Ting Huang
- 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 300192, China
| | - 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 300192, China
| | - Tong Zhu
- 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 300192, 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 300192, 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 300192, China
| | - Wengui Xu
- 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 300060, 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 300192, China
| |
Collapse
|
14
|
Du Y, Xu J, Zheng X, Dang Z, Zhu N, Jiang Z, Li J, Zhu S. NIR-II Protein-Escaping Dyes Enable High-Contrast and Long-Term Prognosis Evaluation of Flap Transplantation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311515. [PMID: 38153348 DOI: 10.1002/adma.202311515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/06/2023] [Indexed: 12/29/2023]
Abstract
Real-time vascular positioning, postoperative flap monitoring, and vascular reconstruction assessment are of great importance in flap transplantation. Cyanine dyes offer the advantage of high resolution in the Near-infrared-II (NIR-II) imaging window. However, the nonspecific binding of many cyanine dyes to endogenous albumin leads to high organ accumulation and skin absorption, resulting in low-quality imaging and poor reproducibility of contrast during long-term (e.g., 7 days) postoperative monitoring. Here, a novel strategy is proposed that can be widely applied to prevent protein binding for NIR-I/II Cl-containing cyanine dyes. This strategy produces protein-escaping dyes, ensuring high fluorescence enhancement in the blood with rapid clearance and no residual fluorescence, allowing for short-term repeatable injections for vascular imaging. This strategy in the perioperative monitoring of pedicle perforator flap models in mice and rats is successfully applied. Furthermore, leveraging the universality of this strategy, multiple nonoverlapping protein-escaping probes that achieve dual-excitation (808 and 1064 nm) interference-free imaging of nerve-vessel and tumor-vessel simultaneously are designed and synthesized. These protein-escaping dyes enable long-term repeatable dual-color imaging of tumor localization, resection, and tumor-vessel reconstruction at the wound site.
Collapse
Affiliation(s)
- Yijing Du
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Jiajun Xu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Xue Zheng
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Zetao Dang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Ningning Zhu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Zijian Jiang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Jia Li
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| |
Collapse
|
15
|
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
|
16
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
17
|
Echigo H, Mishiro K, Munekane M, Fuchigami T, Washiyama K, Takahashi K, Kitamura Y, Wakabayashi H, Kinuya S, Ogawa K. Development of probes for radiotheranostics with albumin binding moiety to increase the therapeutic effects of astatine-211 ( 211At). Eur J Nucl Med Mol Imaging 2024; 51:412-421. [PMID: 37819452 DOI: 10.1007/s00259-023-06457-0] [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: 07/01/2023] [Accepted: 09/23/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE We have developed probes for multiradionuclides radiotheranostics using RGD peptide ([67Ga]Ga-DOTA-c[RGDf(4-I)K] ([67Ga]1) and Ga-DOTA-[211At]c[RGDf(4-At)K] ([211At]2)) for clinical applications. The introduction of an albumin binding moiety (ABM), such as 4-(4-iodophenyl)-butyric acid (IPBA), that has high affinity with the blood albumin and prolongs the circulation half-life can improve the pharmacokinetics of drugs. To perform more effective targeted alpha therapy (TAT), we designed and synthesized Ga-DOTA-K([211At]APBA)-c(RGDfK) ([211At]5) with 4-(4-astatophenyl)-butyric acid (APBA), which has an astato group instead of an iodo group in IPBA. We evaluated whether APBA functions as ABM and [211At]5 is effective for TAT. In addition, we prepared 67Ga-labeled RGD peptide without ABM, [67Ga]Ga-DOTA-K-c(RGDfK) ([67Ga]3), and 125I-labeled RGD peptide with ABM, Ga-DOTA-K([125I]IPBA)-c(RGDfK) ([125I]4), to compare with [211At]5. METHODS Biodistribution experiments of [67Ga]3 without ABM, [125I]4 and [211At]5 with ABM were conducted in normal mice and U-87 MG tumor-bearing mice. In addition, two doses of [211At]5 (370 or 925 kBq) were administered to U-87 MG tumor-bearing mice to confirm the therapeutic effects. RESULTS The blood retention of [125I]4 and [211At]5 was remarkably increased compared to [67Ga]3. Also, [125I]4 and [211At]5 showed similar biodistribution and significantly greater tumor accumulation and retention compared to [67Ga]3. In addition, [211At]5 inhibited tumor growth in a dose-dependent manner. CONCLUSION The functionality of APBA as ABM like IPBA, and the usefulness of [211At]5 as the radionuclide therapy agent for TAT was revealed.
Collapse
Affiliation(s)
- Hiroaki Echigo
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Kohshin Washiyama
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yoji Kitamura
- Research Center for Experimental Modeling of Human Disease, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8641, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
18
|
Bauer I, Ilina E, Zharkov T, Grigorieva E, Chinak O, Kupryushkin M, Golyshev V, Mitin D, Chubarov A, Khodyreva S, Dmitrienko E. Self-Penetrating Oligonucleotide Derivatives: Features of Self-Assembly and Interactions with Serum and Intracellular Proteins. Pharmaceutics 2023; 15:2779. [PMID: 38140119 PMCID: PMC10747088 DOI: 10.3390/pharmaceutics15122779] [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: 11/17/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Lipophilic oligonucleotide derivatives are a potent approach to the intracellular delivery of nucleic acids. The binding of these derivatives to serum albumin is a determinant of their fate in the body, as its structure contains several sites of high affinity for hydrophobic compounds. This study focuses on the features of self-association and non-covalent interactions with human serum albumin of novel self-penetrating oligonucleotide derivatives. The study revealed that the introduction of a triazinyl phosphoramidate modification bearing two dodecyl groups at the 3' end region of the oligonucleotide sequence has a negligible effect on its affinity for the complementary sequence. Dynamic light scattering verified that the amphiphilic oligonucleotides under study can self-assemble into micelle-like particles ranging from 8 to 15 nm in size. The oligonucleotides with dodecyl groups form stable complexes with human serum albumin with a dissociation constant of approximately 10-6 M. The oligonucleotide micelles are simultaneously destroyed upon binding to albumin. Using an electrophoretic mobility shift assay and affinity modification, we examined the ability of DNA duplexes containing triazinyl phosphoramidate oligonucleotides to interact with Ku antigen and PARP1, as well as the mutual influence of PARP1 and albumin or Ku antigen and albumin upon interaction with DNA duplexes. These findings, together with the capability of dodecyl-containing derivatives to effectively penetrate different cells, such as HEK293 and T98G, indicate that the oligonucleotides under study can be considered as a platform for the development of therapeutic preparations with a target effect.
Collapse
Affiliation(s)
- Irina Bauer
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ekaterina Ilina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Timofey Zharkov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
| | - Evgeniya Grigorieva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
| | - Olga Chinak
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
| | - Maxim Kupryushkin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
| | - Victor Golyshev
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
| | - Dmitry Mitin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Alexey Chubarov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Svetlana Khodyreva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
| | - Elena Dmitrienko
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (I.B.); (T.Z.); (O.C.); (M.K.); (V.G.); (D.M.); (S.K.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
19
|
Hassan M, Bokhari TH, Lodhi NA, Khosa MK, Usman M. A review of recent advancements in Actinium-225 labeled compounds and biomolecules for therapeutic purposes. Chem Biol Drug Des 2023; 102:1276-1292. [PMID: 37715360 DOI: 10.1111/cbdd.14311] [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: 03/16/2022] [Revised: 06/03/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
In nuclear medicine, cancers that cannot be cured or can only be treated partially by traditional techniques like surgery or chemotherapy are killed by ionizing radiation as a form of therapeutic treatment. Actinium-225 is an alpha-emitting radionuclide that is highly encouraging as a therapeutic approach and more promising for targeted alpha therapy (TAT). Actinium-225 is the best candidate for tumor cells treatment and has physical characteristics such as high (LET) linear energy transfer (150 keV per μm), half-life (t1/2 = 9.92d), and short ranges (400-100 μm) which prevent the damage of normal healthy tissues. The introduction of various new radiopharmaceuticals and radioisotopes has significantly assisted the advancement of nuclear medicine. Ac-225 radiopharmaceuticals continuously demonstrate their potential as targeted alpha therapeutics. 225 Ac-labeled radiopharmaceuticals have confirmed their importance in medical and clinical areas by introducing [225 Ac]Ac-PSMA-617, [225 Ac]Ac-DOTATOC, [225 Ac]Ac-DOTA-substance-P, reported significantly improved response in patients with prostate cancer, neuroendocrine, and glioma, respectively. The development of these radiopharmaceuticals required a suitable buffer, incubation time, optimal pH, and reaction temperature. There is a growing need to standardize quality control (QC) testing techniques such as radiochemical purity (RCP). This review aims to summarize the development of the Ac-225 labeled compounds and biomolecules. The current state of their reported resulting clinical applications is also summarized as well.
Collapse
Affiliation(s)
- Maria Hassan
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Nadeem Ahmed Lodhi
- Isotope Production Division, Pakistan institute of Nuclear Science & Technology (PINSTECH), Islamabad, Pakistan
| | | | - Muhammad Usman
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
20
|
Tao Y, Jakobsson V, Chen X, Zhang J. Exploiting Albumin as a Versatile Carrier for Cancer Theranostics. Acc Chem Res 2023; 56:2403-2415. [PMID: 37625245 DOI: 10.1021/acs.accounts.3c00309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Affiliation(s)
- Yucen Tao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Department of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and 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
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
21
|
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
|
22
|
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: 1] [Impact Index Per Article: 0.5] [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
|
23
|
Boinapally S, Alati S, Jiang Z, Yan Y, Lisok A, Singh R, Lofland G, Minn I, Hobbs RF, Pomper MG, Banerjee SR. Preclinical Evaluation of a New Series of Albumin-Binding 177Lu-Labeled PSMA-Based Low-Molecular-Weight Radiotherapeutics. Molecules 2023; 28:6158. [PMID: 37630410 PMCID: PMC10459686 DOI: 10.3390/molecules28166158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-based low-molecular-weight agents using beta(β)-particle-emitting radiopharmaceuticals is a new treatment paradigm for patients with metastatic castration-resistant prostate cancer. Although results have been encouraging, there is a need to improve the tumor residence time of current PSMA-based radiotherapeutics. Albumin-binding moieties have been used strategically to enhance the tumor uptake and retention of existing PSMA-based investigational agents. Previously, we developed a series of PSMA-based, β-particle-emitting, low-molecular-weight compounds. From this series, 177Lu-L1 was selected as the lead agent because of its reduced off-target radiotoxicity in preclinical studies. The ligand L1 contains a PSMA-targeting Lys-Glu urea moiety with an N-bromobenzyl substituent in the ε-amino group of Lys. Here, we structurally modified 177Lu-L1 to improve tumor targeting using two known albumin-binding moieties, 4-(p-iodophenyl) butyric acid moiety (IPBA) and ibuprofen (IBU), and evaluated the effects of linker length and composition. Six structurally related PSMA-targeting ligands (Alb-L1-Alb-L6) were synthesized based on the structure of 177Lu-L1. The ligands were assessed for in vitro binding affinity and were radiolabeled with 177Lu following standard protocols. All 177Lu-labeled analogs were studied in cell uptake and selected cell efficacy studies. In vivo pharmacokinetics were investigated by conducting tissue biodistribution studies for 177Lu-Alb-L2-177Lu-Alb-L6 (2 h, 24 h, 72 h, and 192 h) in male NSG mice bearing human PSMA+ PC3 PIP and PSMA- PC3 flu xenografts. Preliminary therapeutic ratios of the agents were estimated from the area under the curve (AUC0-192h) of the tumors, blood, and kidney uptake values. Compounds were obtained in >98% radiochemical yields and >99% purity. PSMA inhibition constants (Kis) of the ligands were in the ≤10 nM range. The long-linker-based agents, 177Lu-Alb-L4 and 177Lu-Alb-L5, displayed significantly higher tumor uptake and retention (p < 0.001) than the short-linker-bearing 177Lu-Alb-L2 and 177Lu-Alb-L3 and a long polyethylene glycol (PEG) linker-bearing agent, 177Lu-Alb-L6. The area under the curve (AUC0-192h) of the PSMA+ PC3 PIP tumor uptake of 177Lu-Alb-L4 and 177Lu-Alb-L5 were >4-fold higher than 177Lu-Alb-L2, 177Lu-Alb-L3, and 177Lu-Alb-L6, respectively. Also, the PSMA+ PIP tumor uptake (AUC0-192h) of 177Lu-Alb-L2 and 177Lu-Alb-L3 was ~1.5-fold higher than 177Lu-Alb-L6. However, the lowest blood AUC0-192h and kidney AUC0-192h were associated with 177Lu-Alb-L6 from the series. Consequently, 177Lu-Alb-L6 displayed the highest ratios of AUC(tumor)-to-AUC(blood) and AUC(tumor)-to-AUC(kidney) values from the series. Among the other agents, 177Lu-Alb-L4 demonstrated a nearly similar ratio of AUC(tumor)-to-AUC(blood) as 177Lu-Alb-L6. The tumor-to-blood ratio was the dose-limiting therapeutic ratio for all of the compounds. Conclusions: 177Lu-Alb-L4 and 177Lu-Alb-L6 showed high tumor uptake in PSMA+ tumors and tumor-to-blood ratios. The data suggest that linker length and composition can be modulated to generate an optimized therapeutic agent.
Collapse
Affiliation(s)
- Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Zirui Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Alla Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Gabriela Lofland
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Robert F. Hobbs
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, 1550 Orleans Street, Cancer Research Building 2, Baltimore, MD 21287, USA; (S.B.); (S.A.); (Z.J.); (I.M.); (R.F.H.); (M.G.P.)
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
24
|
Gaonkar RH, Schmidt YT, Mansi R, Almeida-Hernanadez Y, Sanchez-Garcia E, Harms M, Münch J, Fani M. Development of a New Class of CXCR4-Targeting Radioligands Based on the Endogenous Antagonist EPI-X4 for Oncological Applications. J Med Chem 2023. [PMID: 37328158 DOI: 10.1021/acs.jmedchem.3c00131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The peptide fragment of human serum albumin that was identified as an inhibitor of C-X-C motif chemokine receptor 4 (CXCR4), termed EPI-X4, was investigated as a scaffold for the development of CXCR4-targeting radio-theragnostics. Derivatives of its truncated version JM#21 (ILRWSRKLPCVS) were conjugated to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and tested in Jurkat and Ghost-CXCR4 cells. Ligand-1, -2, -5, -6, -7, -8, and -9 were selected for radiolabeling. Molecular modeling indicated that 177Lu-DOTA incorporation C-terminally did not interfere with the CXCR4 binding. Lipophilicity, in vitro plasma stability, and cellular uptake hinted 177Lu-7 as superior. In Jurkat xenografts, all radioligands showed >90% washout from the body within an hour, with the exception of 177Lu-7 and 177Lu-9. 177Lu-7 demonstrated best CXCR4-tumor targeting. Ex vivo biodistribution and single-photon emission computed tomography (SPECT)/positron emission tomography (PET)/CT imaging of 177Lu-7/68Ga-7 showed the same distribution profile for both radioligands, characterized by very low uptake in all nontargeted organs except the kidneys. The data support the feasibility of CXCR4-targeting with EPI-X4-based radioligands and designate ligand-7 as a lead candidate for further optimization.
Collapse
Affiliation(s)
- Raghuvir Haridas Gaonkar
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| | - Yannik Tim Schmidt
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| | - Yasser Almeida-Hernanadez
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45117, Germany
- Computational Bioengineering, Faculty of Bio- and Chemical Engineering, Technical University Dortmund, Dortmund 44227, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45117, Germany
- Computational Bioengineering, Faculty of Bio- and Chemical Engineering, Technical University Dortmund, Dortmund 44227, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm 89081, Germany
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| |
Collapse
|
25
|
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: 8] [Impact Index Per Article: 4.0] [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
|
26
|
Kim MH, Lee K, Oh K, Kim CH, Kil HS, Lee YJ, Lee KC, Chi DY. Evaluation of PSMA target diagnostic PET tracers for therapeutic monitoring of [ 177Lu]ludotadipep of prostate cancer: Screening of PSMA target efficiency and biodistribution using [ 18F]DCFPyL and [ 68Ga]PSMA-11. Biochem Biophys Res Commun 2023; 651:107-113. [PMID: 36801611 DOI: 10.1016/j.bbrc.2023.02.003] [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: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023]
Abstract
We have compared the similarity of the in vivo distribution of the prostate-specific membrane antigen (PSMA)-targeted positron emission tomography (PET) imaging agents [18F]DCFPyL, [68Ga]galdotadipep, and [68Ga]PSMA-11. This study is designed for a further selection of a PSMA-targeted PET imaging agent for the therapeutic evaluation of [177Lu]ludotadipep, our previously developed prostate-specific membrane antigen (PSMA)-targeted prostate cancer therapeutic radiopharmaceutical. In vitro cell uptake was performed to evaluate the affinity to PSMA using PSMA + PC3-PIP, and PSMA- PC3-flu was used for the study. MicroPET/CT 60 min dynamic imaging and biodistribution were performed at 1, 2, and 4 h after injection. Autoradiography and immunohistochemistry were performed to evaluate the PSMA + tumor target efficiency. In the microPET/CT image, [68Ga]PSMA-11 showed the highest uptake in the kidney among all three compounds. [18F]DCFPyL and [68Ga]PSMA-11 showed similar patterns of in vivo biodistribution and high tumor targeting efficiency, similar to those of[68Ga]galdotadipep. All three agents showed high uptake in tumor tissue on autoradiography, and PSMA expression was confirmed by immunohistochemistry. Thus, [18F]DCFPyL or [68Ga]PSMA-11 can be used as a PET imaging agent to monitor [177Lu]ludotadipep therapy in prostate cancer patients.
Collapse
Affiliation(s)
- Min Hwan Kim
- Research Institute of Radiopharmaceuticals, FutureChem Co. Ltd, Seoul, 04793, Republic of Korea
| | - Kyongkyu Lee
- Research Institute of Radiopharmaceuticals, FutureChem Co. Ltd, Seoul, 04793, Republic of Korea
| | - Keumrok Oh
- Research Institute of Radiopharmaceuticals, FutureChem Co. Ltd, Seoul, 04793, Republic of Korea
| | - Chul Hee Kim
- Research Institute of Radiopharmaceuticals, FutureChem Co. Ltd, Seoul, 04793, Republic of Korea
| | - Hee Seup Kil
- Research Institute of Radiopharmaceuticals, FutureChem Co. Ltd, Seoul, 04793, Republic of Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Republic of Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Republic of Korea
| | - Dae Yoon Chi
- Research Institute of Radiopharmaceuticals, FutureChem Co. Ltd, Seoul, 04793, Republic of Korea.
| |
Collapse
|
27
|
Mishiro K, Imai S, Ematsu Y, Hirose K, Fuchigami T, Munekane M, Kinuya S, Ogawa K. RGD Peptide-Conjugated Dodecaborate with the Ga-DOTA Complex: A Preliminary Study for the Development of Theranostic Agents for Boron Neutron Capture Therapy and Its Companion Diagnostics. J Med Chem 2022; 65:16741-16753. [PMID: 36512639 DOI: 10.1021/acs.jmedchem.2c01586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A boron neutron capture therapy (BNCT) system, using boron-10-introduced agents coupled with companion diagnostics, is anticipated as a promising cancer theranostic. Thus, this study aimed to synthesize and evaluate a probe closo-dodecaborate-(Ga-DOTA)-c(RGDfK) (16) [Ga = gallium, DOTA =1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid, and c(RGDfK) = cyclo(arginine-glycine-aspartate-d-phenylalanine-lysine] containing closo-dodecaborate ([B12H12]2-) as a boron cluster, a [67Ga]Ga-DOTA derivative for nuclear medicine imaging, and an RGD peptide for tumor targeting. Moreover, we prepared a radioiodinated probe [125I]17 in which I-125 is introduced into a closo-dodecaborate moiety of 16. [67Ga]16 and [125I]17 showed high stability and high uptake in cancer cells in vitro. Biodistribution experiments in tumor-bearing mice revealed similar biodistribution patterns between [67Ga]16 and [125I]17, such as a high uptake in the tumor and a low uptake in other non-target tissues. Meanwhile, [125I]17 exhibited higher accumulation in most tissues, including the tumor, than [67Ga]16, probably because of higher albumin binding. The higher the [125I]17 accumulation in the tumor, the more desirable it is for BNCT, with the possibility that the iodo-closo-dodecaborate site may work as an albumin binder.
Collapse
Affiliation(s)
- Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa920-1192, Japan
| | - Sayaka Imai
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa920-1192, Japan
| | - Yuki Ematsu
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa920-1192, Japan
| | - Katsumi Hirose
- Southern Tohoku BNCT Research Center, Koriyama963-8052, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa920-1192, Japan
| | - Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa920-1192, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa920-8641, Japan
| | - Kazuma Ogawa
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa920-1192, Japan.,Graduate School of Medical Sciences, Kanazawa University, Kanazawa920-1192, Japan
| |
Collapse
|
28
|
Radiometal-theranostics: the first 20 years*. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AbstractThis review describes the basic principles of radiometal-theranostics and its dawn based on the development of the positron-emitting 86Y and 86Y-labeled radiopharmaceuticals to quantify biodistribution and dosimetry of 90Y-labeled analogue therapeutics. The nuclear and inorganic development of 86Y (including nuclear and cross section data, irradiation, radiochemical separation and recovery) led to preclinical and clinical evaluation of 86Y-labeled citrate and EDTMP complexes and yielded organ radiation doses in terms of mGy/MBq 90Y. The approach was extended to [86/90Y]Y-DOTA-TOC, yielding again yielded organ radiation doses in terms of mGy/MBq 90Y. The review further discusses the consequences of this early development in terms of further radiometals that were used (68Ga, 177Lu etc.), more chelators that were developed, new biological targets that were addressed (SSTR, PSMA, FAP, etc.) and subsequent generations of radiometal-theranostics that resulted out of that.
Collapse
|
29
|
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] [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
|
30
|
Lindsley C, Müller CE, Bongarzone S. Diagnostic and Therapeutic Radiopharmaceuticals. ACS Pharmacol Transl Sci 2022; 5:835-837. [PMID: 36268118 PMCID: PMC9578137 DOI: 10.1021/acsptsci.2c00173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Craig
W. Lindsley
- Department
of Pharmacology, Department of Chemistry, and Vanderbilt Institute
of Chemical Biology, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Christa E. Müller
- PharmaCenter
Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität Bonn, D-53121 Bonn, Germany
| | - Salvatore Bongarzone
- Technical
Research and Development, Advanced Accelerator
Applications, a Novartis Company, via Ribes 5, Colleretto
Giacosa 10010, Italy
| |
Collapse
|
31
|
Nakashima K, Iikuni S, Watanabe H, Ono M. Application of the Chelator-Based Clickable Radiotheranostic Platform to Moderate-Molecular-Weight Ligands. ACS Med Chem Lett 2022; 13:1642-1647. [PMID: 36262405 PMCID: PMC9575180 DOI: 10.1021/acsmedchemlett.2c00320] [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: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022] Open
Abstract
We have reported that the chelator-based clickable radiotheranostic platform, ADIBO-DOTADG-ALB (ADA), has favorable properties as a radiotheranostic platform for low-molecular-weight ligands. In this study, we evaluated the applicability of ADA to moderate-molecular-weight ligands to expand the utility of the ADA platform. As a moderate-molecular-weight ligand, we selected exendin-4, a peptide-based agonist to glucagon-like peptide-1 receptor (GLP-1R). An exendin-4-incorporated ADA derivative, exendin-4-Cys40-triazole-DOTADG-ALB (EtDA), was radiolabeled with 111In by the conjugation of exendin-4-Cys40 azide to [111In]In-ADA. The click ligation of exendin-4-Cys40 azide to [111In]In-ADA was quantitatively completed in 10 min under ambient conditions. In the in vitro cell-binding assay and albumin-binding assay, [111In]In-EtDA showed strong binding to both a GLP-1R-expressing cell and albumin. In the biodistribution assay, [111In]In-EtDA showed markedly protracted tumor uptake, which was significantly decreased by the coinjection of exendin-4-Cys40. The single photon emission computed tomography (SPECT) image of [111In]In-EtDA visualized the tumor clearly. These results indicated the utility of [111In]In-EtDA as a radiotheranostic agent, suggesting the applicability of the ADA platform to moderate-molecular-weight ligands.
Collapse
Affiliation(s)
- 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
| | - Shimpei Iikuni
- 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
|
32
|
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
|
33
|
Lindsley CW, Müller CE, Bongarzone S. Diagnostic and Therapeutic Radiopharmaceuticals. J Med Chem 2022; 65:12497-12499. [DOI: 10.1021/acs.jmedchem.2c01403] [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)
- Craig W. Lindsley
- Department of Pharmacology, Department of Chemistry, and Vanderbilt Institute of Chemical Biology, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität Bonn, D-53121 Bonn, Germany
| | - Salvatore Bongarzone
- Technical Research and Development, Advanced Accelerator Applications, a Novartis Company, via Ribes 5, Colleretto Giacosa 10010, Italy
| |
Collapse
|
34
|
Lau J, Lee H, Rousseau J, Bénard F, Lin KS. Application of Cleavable Linkers to Improve Therapeutic Index of Radioligand Therapies. Molecules 2022; 27:molecules27154959. [PMID: 35956909 PMCID: PMC9370263 DOI: 10.3390/molecules27154959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 01/11/2023] Open
Abstract
Radioligand therapy (RLT) is an emergent drug class for cancer treatment. The dose administered to cancer patients is constrained by the radiation exposure to normal tissues to maintain an appropriate therapeutic index. When a radiopharmaceutical or its radiometabolite is retained in the kidneys, radiation dose deposition in the kidneys can become a dose-limiting factor. A good exemplar is [177Lu]Lu-DOTATATE, where patients receive a co-infusion of basic amino acids for nephroprotection. Besides peptides, there are other classes of targeting vectors like antibody fragments, antibody mimetics, peptidomimetics, and small molecules that clear through the renal pathway. In this review, we will review established and emerging strategies that can be used to mitigate radiation-induced nephrotoxicity, with a focus on the development and incorporation of cleavable linkers for radiopharmaceutical designs. Finally, we offer our perspectives on cleavable linkers for RLT, highlighting future areas of research that will help advance the technology.
Collapse
Affiliation(s)
- Joseph Lau
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Hwan Lee
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Julie Rousseau
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Correspondence: ; Tel.: +1-604-675-8208
| |
Collapse
|
35
|
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
|
36
|
Korde A, Mikolajczak R, Kolenc P, Bouziotis P, Westin H, Lauritzen M, Koole M, Herth MM, Bardiès M, Martins AF, Paulo A, Lyashchenko SK, Todde S, Nag S, Lamprou E, Abrunhosa A, Giammarile F, Decristoforo C. Practical considerations for navigating the regulatory landscape of non-clinical studies for clinical translation of radiopharmaceuticals. EJNMMI Radiopharm Chem 2022; 7:18. [PMID: 35852679 PMCID: PMC9296747 DOI: 10.1186/s41181-022-00168-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background The development of radiopharmaceuticals requires extensive evaluation before they can be applied in a diagnostic or therapeutic setting in Nuclear Medicine. Chemical, radiochemical, and pharmaceutical parameters must be established and verified to ensure the quality of these novel products.
Main body To provide supportive evidence for the expected human in vivo behaviour, particularly related to safety and efficacy, additional tests, often referred to as “non-clinical” or “preclinical” are mandatory. This document is an outcome of a Technical Meeting of the International Atomic Energy Agency. It summarises the considerations necessary for non-clinical studies to accommodate the regulatory requirements for clinical translation of radiopharmaceuticals. These considerations include non-clinical pharmacology, radiation exposure and effects, toxicological studies, pharmacokinetic modelling, and imaging studies. Additionally, standardisation of different specific clinical applications is discussed.
Conclusion This document is intended as a guide for radiopharmaceutical scientists, Nuclear Medicine specialists, and regulatory professionals to bring innovative diagnostic and therapeutic radiopharmaceuticals into the clinical evaluation process in a safe and effective way.
Collapse
Affiliation(s)
- Aruna Korde
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna International Centre, PO Box 100, 1400, Vienna, Austria
| | - Renata Mikolajczak
- Radioisotope Centre POLATOM, National Centre for Nuclear Research, Andrzej Soltan 7, 05-400, Otwock, Poland
| | - Petra Kolenc
- Department of Nuclear Medicine, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Penelope Bouziotis
- National Centre for Scientific Research "Demokritos", Institute of Nuclear & Radiological Sciences and Technology, Energy & Safety, 15341, Athens, Greece
| | - Hadis Westin
- Department of Immunology, Genetics and Pathology, Ridgeview Instruments AB, Uppsala Universitet, Dag Hammarskjölds Väg 36A, 752 37, Uppsala, Sweden
| | - Mette Lauritzen
- Bruker BioSpin MRI GmbH, Rudolf-Plank-Str. 23, 76275, Ettlingen, Germany
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Katholieke Universiteit Leuven, 3000, Louvain, Belgium
| | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Manuel Bardiès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Institut Régional du Cancer de Montpellier (ICM), Université de Montpellier, 34298, Montpellier, France
| | - Andre F Martins
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tübingen, Röntgenweg 13/1, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Antonio Paulo
- Centro de Ciências E Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela Lrs, Campus Tecnológico e Nuclear, Estrada Nacional 10, Km 139.7, 2695-066, Lisbon, Portugal
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio Todde
- Department of Medicine and Surgery, University of Milano-Bicocca, Tecnomed Foundation, Milan, Italy
| | - Sangram Nag
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 76, Stockholm, Sweden
| | - Efthimis Lamprou
- Bioemtech, Lefkippos Attica Technology Park-N.C.S.R Demokritos, Athens, Greece
| | - Antero Abrunhosa
- ICNAS/CIBIT, Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - Francesco Giammarile
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna International Centre, PO Box 100, 1400, Vienna, Austria
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
37
|
Iikuni S, Ohara T, Watanabe H, Ono M. Structure-Activity Relationships and Pharmacokinetics of 111In-Labeled Glucagon-like Peptide-1 Receptor-Targeting Exendin-4 Derivatives Conjugated with Albumin Binder Moieties. Mol Pharm 2022; 19:2832-2839. [PMID: 35757958 DOI: 10.1021/acs.molpharmaceut.2c00201] [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: 11/29/2022]
Abstract
Insulinomas are neuroendocrine tumors that are derived from pancreatic β-cells, and they often overexpress the glucagon-like peptide-1 receptor (GLP-1R). Radiolabeled exendin-4 derivatives have been used to noninvasively detect the GLP-1R during the diagnosis and preoperative localization of insulinomas; however, their marked renal accumulation can hinder the imaging of pancreatic tail lesions. In this study, we designed and synthesized 111In-labeled exendin-4 derivatives that possessed 4-(4-substituted phenyl)-moieties as albumin binder (ALB) moieties ([111In]In-E4DA2-4), and studied their structure-activity relationships and pharmacokinetics (as well as those of [111In]In-E4DA1, which we previously reported) to determine their usefulness as radioligands for GLP-1R imaging. 111In-labeling was performed by reacting maleimide precursors with [111In]InCl3 in 2-(N-morpholino)ethanesulfonic acid buffer, and then, the products were conjugated with exendin-4-Cys40. A saturation binding assay using GLP-1R-expressing INS-1 cells was carried out to evaluate the in vitro affinity of the radioligands for the cells. In addition, the affinity of the 111In-labeled derivatives for human serum albumin (HSA) was evaluated in an HSA-binding assay. Furthermore, an in vivo biodistribution study and single-photon emission computed tomography (SPECT) imaging were performed using INS-1 tumor-bearing mice. [111In]In-E4DA1-4 were prepared at radiochemical yields of 6-17%. In the saturation binding assay, [111In]In-E4DA1-4 showed a similar affinity for the INS-1 cells, indicating that the kind of ALB moiety used had no effect on the affinity of the exendin-4 derivatives for the cells. In the HSA-binding assay, [111In]In-E4DA1-4 all bound to HSA. In the biodistribution assay, [111In]In-E4DA1-4 exhibited marked tumor accumulation and retention. In addition, they showed lower renal accumulation than previously reported exendin-4-based radioligands without ALB moieties. The pharmacokinetics of the 111In-labeled exendin-4 derivatives varied markedly according to the kind of ALB moiety used. In particular, [111In]In-E4DA2, which contained a 4-(4-bromophenyl)butyric acid derivative as an ALB moiety, showed the highest tumor accumulation. SPECT imaging with [111In]In-E4DA2 clearly visualized INS-1 tumors with no marked accumulation in normal organs. These results provide important information that will aid the design of novel exendin-4-based radioligands targeting the GLP-1R.
Collapse
Affiliation(s)
- Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takaki Ohara
- 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
|
38
|
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
|
39
|
Palmitic Acid-Conjugated Radiopharmaceutical for Integrin αvβ3-Targeted Radionuclide Therapy. Pharmaceutics 2022; 14:pharmaceutics14071327. [PMID: 35890224 PMCID: PMC9321335 DOI: 10.3390/pharmaceutics14071327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is an emerging approach for patients with unresectable or metastatic tumors. Our previously optimized RGD peptide (3PRGD2) has excellent targeting specificity for a variety of integrin αvβ3/αvβ5-positive tumors and has been labeled with the therapeutic radionuclide [177Lu]LuCl3 for targeted radiotherapy of tumors. However, the rapid clearance of [177Lu]Lu-DOTA-3PRGD2 (177Lu-3PRGD2) in vivo requires two doses of 111 MBq/3 mCi to achieve effective tumor suppression, limiting its further clinical application. Albumin binders have been attached to drugs to facilitate binding to albumin in vivo to prolong the drug half-life in plasma and obtain long-term effects. In this study, we modified 3PRGD2 with albumin-binding palmitic acid (Palm-3PRGD2) and then radiolabeled Palm-3PRGD2 with 177Lu. [177Lu]Lu-DOTA-Palm-3PRGD2 (177Lu-Palm-3PRGD2) retained a specific binding affinity for integrin αvβ3/αvβ5, with an IC50 value of 5.13 ± 1.16 nM. Compared with 177Lu-3PRGD2, the 177Lu-Palm-3PRGD2 circulation time in blood was more than 6 times longer (slow half-life: 73.42 min versus 11.81 min), and the tumor uptake increased more than fivefold (21.34 ± 4.65 %IA/g and 4.11 ± 0.70 %IA/g at 12 h post-injection). Thus, the significant increase in tumor uptake and tumor retention resulted in enhanced efficacy of targeted radiotherapy, and tumor growth was completely inhibited by a single and relatively lowdose of 18.5 MBq/0.5 mCi. Thus, 177Lu-Palm-3PRGD2 shows great potential for clinical application.
Collapse
|
40
|
Meng L, Fang J, Zhao L, Wang T, Yuan P, Zhao Z, Zhuang R, Lin Q, Chen H, Chen X, Zhang X, Guo Z. Rational Design and Pharmacomodulation of Protein-Binding Theranostic Radioligands for Targeting the Fibroblast Activation Protein. J Med Chem 2022; 65:8245-8257. [PMID: 35658448 DOI: 10.1021/acs.jmedchem.1c02162] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The fibroblast activation protein (FAP), overexpressed on cancer-associated fibroblasts (CAFs), has become a valuable target for tumor diagnosis and therapy. However, most FAP-based radioligands show insufficient tumor uptake and retention. In this study, three novel albumin-binding FAP ligands (denoted as FSDD0I, FSDD1I, and FSDD3I) were labeled with 68Ga and 177Lu to overcome these limitations. Cell-based studies and molecular docking assays were performed to identify the specificity and protein-binding properties for FAP. Positron emission tomography (PET) scans in human hepatocellular carcinoma patient-derived xenografts (HCC-PDXs) animal models revealed longer blood retention of 68Ga-FSDD0I than 68Ga-FAPI-04, 68Ga-FSDD1I, and 68Ga-FSDD3I. Remarkably, 68Ga-FSDD3I had prominent tumor-to-nontarget (T/NT) ratios. The prominent tumor retention properties of 177Lu-FSDD0I in single photon emission computed tomography (SPECT) imaging and biodistribution studies were demonstrated. In summary, this study reports a proof-of-concept study of albumin-binding radioligands for FAP-targeted imaging and targeted radionuclide therapy (TRT).
Collapse
Affiliation(s)
- Lingxin Meng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jianyang Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Liang Zhao
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.,Department of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Tingting Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Pu Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zuoquan Zhao
- Department of Nuclear Medicine, Cardiovascular Institute and FuWai Hospital, Chinese Academy of Medical Sciences, Beijing 100037, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qin Lin
- Department of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Haojun Chen
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, 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.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
41
|
Xu M, Zhang P, Ding J, Chen J, Huo L, Liu Z. Albumin Binder-Conjugated Fibroblast Activation Protein Inhibitor Radiopharmaceuticals for Cancer Therapy. J Nucl Med 2022; 63:952-958. [PMID: 34593598 PMCID: PMC9157728 DOI: 10.2967/jnumed.121.262533] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/25/2021] [Indexed: 11/16/2022] Open
Abstract
Fibroblast activation protein (FAP) has become an attractive target for diagnosis and therapy, and a series of FAP inhibitor (FAPI)-based radiotracers has been developed and had excellent performance for diagnosis outcomes in clinical applications. Yet, their fast clearance and insufficient tumor retention have hampered their further clinical application in cancer treatment. In this study, we developed 2 albumin binder-conjugated FAPI radiotracers, TEFAPI-06 and TEFAPI-07. They were derived from FAPI-04 and were optimized by conjugating 2 types of well-studied albumin binders, 4-(p-iodophenyl) butyric acid moiety (TEFAPI-06) and truncated Evans blue moiety (TEFAPI-07), to try to overcome the above limitations at the expense of prolonging the blood circulation. Methods: TEFAPI-06 and TEFAPI-07 were synthesized and labeled with 68Ga, 86Y, and 177Lu successfully. A series of cell assays was performed to identify the binding affinity and FAP specificity in vitro. PET imaging, SPECT imaging, and biodistribution studies were performed to evaluate the pharmacokinetics in pancreatic cancer patient-derived xenograft (PDX) animal models. The cancer treatment efficacy of 177Lu-TEFAPI-06 and 177Lu-TEFAPI-07 were evaluated in pancreatic cancer PDX-bearing mice. Results: The binding affinities (dissociation constants) to FAP of 68Ga-TEFAPI-06 and 68Ga-TEFAPI-07 were 10.16 ± 2.56 nM and 7.81 ± 2.28 nM, respectively, which were comparable with that of 68Ga-FAPI-04. Comparative PET imaging of HT-1080-FAP and HT-1080 tumor-bearing mice and a blocking study showed the FAP-targeting ability in vivo of these 2 tracers. Compared with 177Lu-FAPI-04, PET imaging, SPECT imaging, and biodistribution studies of TEFAPI-06 and TEFAPI-07 demonstrated their remarkably enhanced tumor accumulation and retention, respectively. Notable tumor growth inhibition by 177Lu-TEFAPI-06 and 177Lu-TEFAPI-07 were observed, whereas the control group and the group treated by 177Lu-FAPI-04 showed a slight therapeutic effect. Conclusion: Two albumin binder-conjugated FAPI radiopharmaceuticals have been developed and evaluated in vitro and in vivo. Significantly improved tumor uptake and retention were observed, compared with the original FAPI tracer. Both 177Lu-TEFAPI-06 and 177Lu-TEFAPI-07 showed remarkable growth inhibition of PDX tumors, whereas the side effects were almost negligible, demonstrating that these radiopharmaceuticals are promising for further clinical translational studies.
Collapse
Affiliation(s)
- Mengxin Xu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Pu Zhang
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Jie Ding
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China; and
| | - Junyi Chen
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China; and
| | - Zhibo Liu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China;
- Peking University-Tsinghua University Center for Life Sciences, Beijing, China
| |
Collapse
|
42
|
Feasibility of Gd-Based prostate cancer targeted magnetic resonance agents using prostate specific membrane antigen. Biochem Biophys Res Commun 2022; 607:152-157. [DOI: 10.1016/j.bbrc.2022.03.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/25/2022] [Indexed: 11/19/2022]
|
43
|
Borgna F, Deberle LM, Busslinger SD, Tschan VJ, Walde LM, Becker AE, Schibli R, Müller C. Preclinical Investigations to Explore the Difference between the Diastereomers [ 177Lu]Lu-SibuDAB and [ 177Lu]Lu-RibuDAB toward Prostate Cancer Therapy. Mol Pharm 2022; 19:2105-2114. [PMID: 35544699 DOI: 10.1021/acs.molpharmaceut.1c00994] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
[177Lu]Lu-Ibu-DAB-PSMA, a radioligand modified with ibuprofen as the albumin binder, showed higher accumulation in PSMA-positive tumors of mice than the clinically used [177Lu]Lu-PSMA-617 but lower retention in non-targeted tissues than previously developed albumin-binding PSMA radioligands. The aim of this study was to investigate whether the stereochemistry of the incorporated ibuprofen affects the radioligand's in vitro and in vivo properties and to select the more favorable radioligand for further development. For this purpose, SibuDAB and RibuDAB containing (S)- and (R)-ibuprofen, respectively, were synthesized and labeled with lutetium-177. In vitro, the two isomers had similar properties; however, [177Lu]Lu-SibuDAB showed increased binding to mouse and human plasma proteins (91 ± 1 and 88 ± 2%, respectively) compared to [177Lu]Lu-RibuDAB (75 ± 2 and 79 ± 2%, respectively). In vivo, [177Lu]Lu-SibuDAB was metabolically more stable than [177Lu]Lu-RibuDAB with ∼90 vs ∼67% intact radioligand detected in the blood at 4 h post injection (p.i.). In line with the lower albumin-binding affinity, the blood clearance of [177Lu]Lu-RibuDAB in mice was considerably faster [27% of injected activity (% IA), 1 h p.i.] than for [177Lu]Lu-SibuDAB (50% IA, 1 h p.i.). Time-dependent biodistribution studies performed in tumor-bearing athymic nude mice showed high PSMA-specific tumor uptake for both isomers. A twofold increased area under the curve (AUC0→8d) of the blood retention was determined for [177Lu]Lu-SibuDAB as compared to [177Lu]Lu-RibuDAB, whereas the kidney AUC0→8d value of [177Lu]Lu-SibuDAB was only half as high as for [177Lu]Lu-RibuDAB. As a result, a more favorable tumor-to-kidney AUC0→8d ratio was obtained for [177Lu]Lu-SibuDAB, which was also visualized on SPECT/CT images. Based on its improved kidney clearance and higher metabolic stability, [177Lu]Lu-SibuDAB was selected as the more favorable radioligand. Therapy studies performed with [177Lu]Lu-SibuDAB (5 MBq/mouse) demonstrated the anticipated therapeutic superiority over the current gold-standard [177Lu]Lu-PSMA-617 (5 MBq/mouse). The significantly increased survival time of mice treated with [177Lu]Lu-SibuDAB as compared to those injected with [177Lu]Lu-PSMA-617 justifies further development of this novel radioligand toward clinical application.
Collapse
Affiliation(s)
- Francesca Borgna
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland
| | - Luisa M Deberle
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland
| | - Viviane J Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland
| | - Laura M Walde
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland
| | - Anna E Becker
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich 8093, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen-PSI 5232, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
44
|
Gao W, Hu H, Dai L, He M, Yuan H, Zhang H, Liao J, Wen B, Li Y, Palmisano M, Traore MDM, Zhou S, Sun D. Structure‒tissue exposure/selectivity relationship (STR) correlates with clinical efficacy/safety. Acta Pharm Sin B 2022; 12:2462-2478. [PMID: 35646532 PMCID: PMC9136610 DOI: 10.1016/j.apsb.2022.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/23/2022] [Accepted: 02/12/2022] [Indexed: 11/17/2022] Open
Abstract
Drug optimization, which improves drug potency/specificity by structure‒activity relationship (SAR) and drug-like properties, is rigorously performed to select drug candidates for clinical trials. However, the current drug optimization may overlook the structure‒tissue exposure/selectivity-relationship (STR) in disease-targeted tissues vs. normal tissues, which may mislead the drug candidate selection and impact the balance of clinical efficacy/toxicity. In this study, we investigated the STR in correlation with observed clinical efficacy/toxicity using seven selective estrogen receptor modulators (SERMs) that have similar structures, same molecular target, and similar/different pharmacokinetics. The results showed that drug's plasma exposure was not correlated with drug's exposures in the target tissues (tumor, fat pad, bone, uterus), while tissue exposure/selectivity of SERMs was correlated with clinical efficacy/safety. Slight structure modifications of four SERMs did not change drug's plasma exposure but altered drug's tissue exposure/selectivity. Seven SERMs with high protein binding showed higher accumulation in tumors compared to surrounding normal tissues, which is likely due to tumor EPR effect of protein-bound drugs. These suggest that STR alters drug's tissue exposure/selectivity in disease-targeted tissues vs. normal tissues impacting clinical efficacy/toxicity. Drug optimization needs to balance the SAR and STR in selecting drug candidate for clinical trial to improve success of clinical drug development.
Collapse
Affiliation(s)
- Wei Gao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hongxiang Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lipeng Dai
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Miao He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hebao Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Huixia Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jinhui Liao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yan Li
- Translational Development and Clinical Pharmacology, Bristol Myers Squibb, Summit, NJ 07920, USA
| | - Maria Palmisano
- Translational Development and Clinical Pharmacology, Bristol Myers Squibb, Summit, NJ 07920, USA
| | - Mohamed Dit Mady Traore
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Simon Zhou
- Translational Development and Clinical Pharmacology, Bristol Myers Squibb, Summit, NJ 07920, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
45
|
Mayoral-Peña K, González Peña OI, Orrantia Clark AM, Flores-Vallejo RDC, Oza G, Sharma A, De Donato M. Biorecognition Engineering Technologies for Cancer Diagnosis: A Systematic Literature Review of Non-Conventional and Plausible Sensor Development Methods. Cancers (Basel) 2022; 14:1867. [PMID: 35454775 PMCID: PMC9030888 DOI: 10.3390/cancers14081867] [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: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 12/21/2022] Open
Abstract
Cancer is the second cause of mortality worldwide. Early diagnosis of this multifactorial disease is challenging, especially in populations with limited access to healthcare services. A vast repertoire of cancer biomarkers has been studied to facilitate early diagnosis; particularly, the use of antibodies against these biomarkers has been of interest to detect them through biorecognition. However, there are certain limitations to this approach. Emerging biorecognition engineering technologies are alternative methods to generate molecules and molecule-based scaffolds with similar properties to those presented by antibodies. Molecularly imprinted polymers, recombinant antibodies, and antibody mimetic molecules are three novel technologies commonly used in scientific studies. This review aimed to present the fundamentals of these technologies and address questions about how they are implemented for cancer detection in recent scientific studies. A systematic analysis of the scientific peer-reviewed literature regarding the use of these technologies on cancer detection was carried out starting from the year 2000 up to 2021 to answer these questions. In total, 131 scientific articles indexed in the Web of Science from the last three years were included in this analysis. The results showed that antibody mimetic molecules technology was the biorecognition technology with the highest number of reports. The most studied cancer types were: multiple, breast, leukemia, colorectal, and lung. Electrochemical and optical detection methods were the most frequently used. Finally, the most analyzed biomarkers and cancer entities in the studies were carcinoembryonic antigen, MCF-7 cells, and exosomes. These technologies are emerging tools with adequate performance for developing biosensors useful in cancer detection, which can be used to improve cancer diagnosis in developing countries.
Collapse
Affiliation(s)
- Kalaumari Mayoral-Peña
- School of Engineering and Sciences, Campus Queretaro, Tecnologico de Monterrey, Av. Epigmenio González No. 500, San Pablo, Queretaro 76130, Mexico; (K.M.-P.); (A.S.)
| | - Omar Israel González Peña
- School of Engineering and Sciences, Campus Monterrey, Tecnologico de Monterrey, Av. Eugenio Garza Sada Sur No. 2501, Tecnológico, Monterrey 64849, Mexico
- Institute for the Future of Education, Tecnologico de Monterrey, Av. Eugenio Garza Sada Sur No. 2501, Tecnológico, Monterrey 64849, Mexico
| | - Alexia María Orrantia Clark
- School of Engineering and Sciences, Campus Mexico City, Tecnologico de Monterrey, C. Puente 222, Ejidos de Huipulco, Tlalpan, Mexico City 14380, Mexico;
| | - Rosario del Carmen Flores-Vallejo
- Department of Biomedical Engineering and Mechatronics, Campus Toluca, Universidad del Valle de México (UVM), C. De Las Palmas Poniente 439, San Jorge Pueblo Nuevo, Metepec 52164, Mexico;
| | - Goldie Oza
- Laboratorio Nacional de Micro y Nanofluídica (LABMyN), Centro de Investigación y Desarrollo Tecnológico en Electroquímica (CIDETEQ), Parque San Fandila, Pedro Escobedo, Queretaro 76703, Mexico;
| | - Ashutosh Sharma
- School of Engineering and Sciences, Campus Queretaro, Tecnologico de Monterrey, Av. Epigmenio González No. 500, San Pablo, Queretaro 76130, Mexico; (K.M.-P.); (A.S.)
| | - Marcos De Donato
- School of Engineering and Sciences, Campus Queretaro, Tecnologico de Monterrey, Av. Epigmenio González No. 500, San Pablo, Queretaro 76130, Mexico; (K.M.-P.); (A.S.)
| |
Collapse
|
46
|
Davis RA, Hausner SH, Harris R, Sutcliffe JL. A Comparison of Evans Blue and 4-( p-Iodophenyl)butyryl Albumin Binding Moieties on an Integrin α vβ 6 Binding Peptide. Pharmaceutics 2022; 14:pharmaceutics14040745. [PMID: 35456579 PMCID: PMC9025560 DOI: 10.3390/pharmaceutics14040745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 01/26/2023] Open
Abstract
Serum albumin binding moieties (ABMs) such as the Evans blue (EB) dye fragment and the 4-(p-iodophenyl)butyryl (IP) have been used to improve the pharmacokinetic profile of many radiopharmaceuticals. The goal of this work was to directly compare these two ABMs when conjugated to an integrin αvβ6 binding peptide (αvβ6-BP); a peptide that is currently being used for positron emission tomography (PET) imaging in patients with metastatic cancer. The ABM-modified αvβ6-BP peptides were synthesized with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetracetic acid (DOTA) chelator for radiolabeling with copper-64 to yield [64Cu]Cu DOTA-EB-αvβ6-BP ([64Cu]1) and [64Cu]Cu DOTA-IP-αvβ6-BP ([64Cu]2). Both peptides were evaluated in vitro for serum albumin binding, serum stability, and cell binding and internalization in the paired engineered melanoma cells DX3puroβ6 (αvβ6 +) and DX3puro (αvβ6 −), and pancreatic BxPC-3 (αvβ6 +) cells and in vivo in a BxPC-3 xenograft mouse model. Serum albumin binding for [64Cu]1 and [64Cu]2 was 53−63% and 42−44%, respectively, with good human serum stability (24 h: [64Cu]1 76%, [64Cu]2 90%). Selective αvβ6 cell binding was observed for both [64Cu]1 and [64Cu]2 (αvβ6 (+) cells: 30.3−55.8% and 48.5−60.2%, respectively, vs. αvβ6 (−) cells <3.1% for both). In vivo BxPC-3 tumor uptake for both peptides at 4 h was 5.29 ± 0.59 and 7.60 ± 0.43% ID/g ([64Cu]1 and [64Cu]2, respectively), and remained at 3.32 ± 0.46 and 4.91 ± 1.19% ID/g, respectively, at 72 h, representing a >3-fold improvement over the non-ABM parent peptide and thereby providing improved PET images. Comparing [64Cu]1 and [64Cu]2, the IP-ABM-αvβ6-BP [64Cu]2 displayed higher serum stability, higher tumor accumulation, and lower kidney and liver accumulation, resulting in better tumor-to-organ ratios for high contrast visualization of the αvβ6 (+) tumor by PET imaging.
Collapse
Affiliation(s)
- Ryan A. Davis
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
| | - Sven H. Hausner
- Department of Internal Medicine, Division of Hematology/Oncology, University of California, Davis, CA 95817, USA; (S.H.H.); (R.H.)
| | - Rebecca Harris
- Department of Internal Medicine, Division of Hematology/Oncology, University of California, Davis, CA 95817, USA; (S.H.H.); (R.H.)
| | - Julie L. Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
- Department of Internal Medicine, Division of Hematology/Oncology, University of California, Davis, CA 95817, USA; (S.H.H.); (R.H.)
- Center for Molecular and Genomic Imaging, University of California, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-916-734-5536
| |
Collapse
|
47
|
Harms M, Hansson RF, Carmali S, Almeida-Hernández Y, Sanchez-Garcia E, Münch J, Zelikin AN. Dimerization of the Peptide CXCR4-Antagonist on Macromolecular and Supramolecular Protraction Arms Affords Increased Potency and Enhanced Plasma Stability. Bioconjug Chem 2022; 33:594-607. [PMID: 35293739 DOI: 10.1021/acs.bioconjchem.2c00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peptides are prime drug candidates due to their high specificity of action but are disadvantaged by low proteolytic stability. Here, we focus on the development of stabilized analogues of EPI-X4, an endogenous peptide antagonist of CXCR4. We synthesized macromolecular peptide conjugates and performed side-by-side comparison with their albumin-binding counterparts and considered monovalent conjugates, divalent telechelic conjugates, and Y-shaped peptide dimers. All constructs were tested for competition with the CXCR4 antibody-receptor engagement, inhibition of receptor activation, and inhibition of the CXCR4-tropic human immunodeficiency virus infection. We found that the Y-shaped conjugates were more potent than the parent peptide and at the same time more stable in human plasma, with a favorable outlook for translational studies.
Collapse
Affiliation(s)
- Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rikke Fabech Hansson
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Sheiliza Carmali
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Yasser Almeida-Hernández
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Alexander N Zelikin
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
48
|
Benešová M, Guzik P, Deberle LM, Busslinger SD, Landolt T, Schibli R, Müller C. Design and Evaluation of Novel Albumin-Binding Folate Radioconjugates: Systematic Approach of Varying the Linker Entities. Mol Pharm 2022; 19:963-973. [PMID: 35192367 DOI: 10.1021/acs.molpharmaceut.1c00932] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor targeting using folate radioconjugates is a promising strategy for theragnostics of folate receptor-positive tumors. The aim of this study was to investigate the impact of structural modifications of folate radioconjugates on their pharmacokinetic properties. Four novel folate radioconjugates ([177Lu]Lu-OxFol-2, [177Lu]Lu-OxFol-3, [177Lu]Lu-OxFol-4, and [177Lu]Lu-OxFol-5), modified with a lipophilic or hydrophilic linker entity in close proximity to the albumin-binding 4-(p-iodophenyl)butanoate entity or the DOTA chelator, respectively, were designed and evaluated for comparison with the previously developed [177Lu]Lu-OxFol-1. A hydrophobic 4-(aminomethyl)benzoic acid linker, incorporated in close proximity to the 4-(p-iodophenyl)butanoate entity, enhanced the albumin-binding properties (relative affinity 7.3) of [177Lu]Lu-OxFol-3 as compared to those of [177Lu]Lu-OxFol-1 (relative affinity set as 1.0). On the other hand, a hydrophilic d-glutamic acid (d-Glu) linker entity used in [177Lu]Lu-OxFol-2 compromised the albumin-binding properties. [177Lu]Lu-OxFol-4 and [177Lu]Lu-OxFol-5, in which the respective linker entities were incorporated adjacent to the DOTA chelator, showed similar albumin-binding properties (0.6 and 1.0, respectively) as [177Lu]Lu-OxFol-1. Biodistribution studies in KB tumor-bearing nude mice revealed twofold higher tumor-to-kidney ratios at 4 h and 24 h after injection of [177Lu]Lu-OxFol-3 (∼1.2) than after injection of [177Lu]Lu-OxFol-1 (∼0.6). The tumor-to-kidney ratios of [177Lu]Lu-OxFol-2 were, however, much lower (∼0.2) due to the high kidney retention of this radioconjugate. The tumor-to-kidney ratios of [177Lu]Lu-OxFol-5 were only slightly increased (∼0.9), and the ratios for [177Lu]Lu-OxFol-4 (∼0.7) were in the same range as for [177Lu]Lu-OxFol-1. SPECT/CT imaging studies demonstrated similar tumor uptake of all radioconjugates but a clearly improved tumor-to-kidney ratio for [177Lu]Lu-OxFol-3 as compared to that for [177Lu]Lu-OxFol-1. Based on these data, it can be concluded that the linker entity in close proximity to the 4-(p-iodophenyl)butanoate entity affects the radioconjugate's pharmacokinetic profile considerably due to the altered affinity to albumin. Changes in the linker entity, which connects the DOTA chelator with the folate molecule, do not have a major impact on the radioconjugate's tissue distribution profile, however. As a result of these findings, [177Lu]Lu-OxFol-3 had a comparable therapeutic effect to that of [177Lu]Lu-OxFol-1 but appeared advantageous in preventing kidney damage. Provided that the kidneys will present the dose-limiting organs in patients, [177Lu]Lu-OxFol-3 would be the preferred candidate for a clinical translation.
Collapse
Affiliation(s)
- Martina Benešová
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Patrycja Guzik
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Luisa M Deberle
- 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
| | - Tanja Landolt
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.,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-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
49
|
Iikuni S, Kamei I, Ohara T, Watanabe H, Ono M. Development of an 111In-Labeled Glucagon-Like Peptide-1 Receptor-Targeting Exendin-4 Derivative that Exhibits Reduced Renal Uptake. Mol Pharm 2022; 19:1019-1027. [PMID: 35138111 DOI: 10.1021/acs.molpharmaceut.2c00068] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Insulinomas are neuroendocrine tumors that are mainly found in the pancreas. Surgical resection is currently the first-line treatment for insulinomas; thus, it is vital to preoperatively determine their locations. The marked expression of the glucagon-like peptide-1 receptor (GLP-1R) is seen in pancreatic β-cells and almost all insulinomas. Radiolabeled derivatives of exendin-4, a GLP-1R agonist, have been used with nuclear medicine imaging techniques for the in vivo detection of the GLP-1R; however, their marked renal accumulation can hinder the imaging of pancreatic tail lesions. To develop a GLP-1R imaging probe that exhibits reduced renal accumulation, we designed and synthesized a straight-chain GLP-1R-targeting radioligand, [111In]In-E4DA1, which consisted of exendin-4, DOTADG (a chelator), and an (iodophenyl)butyric acid derivative (an albumin binder [ALB]). We performed preclinical evaluations of [111In]In-E4DA1 to investigate its utility as a GLP-1R imaging probe. [111In]In-E4DA1 and [111In]In-E4D (a control compound lacking the ALB moiety) were prepared by reacting the corresponding precursors with [111In]InCl3 in buffer. Cell-binding and human serum albumin (HSA)-binding assays were performed to assess the in vitro affinity of the molecules for INS-1 (GLP-1R-positive) cells and albumin, respectively. A biodistribution assay and single-photon emission computed tomography imaging were carried out using INS-1 tumor-bearing mice. In the cell-binding assay, [111In]In-E4DA1 and [111In]In-E4D exhibited in vitro binding to INS-1 cells. In the HSA-binding assay, [111In]In-E4DA1 bound to HSA, while [111In]In-E4D showed little HSA binding. The in vivo experiments involving INS-1 tumor-bearing mice revealed that the introduction of an ALB moiety into the DOTADG-based exendin-4 derivative markedly increased the molecule's tumor accumulation while decreasing its renal accumulation. In addition, [111In]In-E4DA1 exhibited greater tumor accumulation than renal accumulation, whereas previously reported radiolabeled exendin-4 derivatives demonstrated much higher accumulation in the kidneys than in tumors. These results indicate that [111In]In-E4DA1 may be a useful GLP-1R imaging probe, as it demonstrates only slight renal accumulation.
Collapse
Affiliation(s)
- Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ichiro Kamei
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Takaki Ohara
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
50
|
Stroet MCM, de Blois E, de Jong M, Seimbille Y, Mezzanotte L, Löwik CWGM, Panth KM. Improved Multimodal Tumor Necrosis Imaging with IRDye800CW-DOTA Conjugated to an Albumin-Binding Domain. Cancers (Basel) 2022; 14:cancers14040861. [PMID: 35205609 PMCID: PMC8870237 DOI: 10.3390/cancers14040861] [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: 12/10/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Anti-tumor treatment efficacy is determined by tumor shrinkage, which takes valuable time to become apparent and poses a risk of unnecessary treatment with severe side effects. Therefore, there is an unmet need for more reliable and specific methods to monitor treatment efficacy. We explore radiolabeled cyanines for imaging tumor necrosis as a unique marker for therapy efficacy. Moreover, spontaneous tumor necrosis is a hallmark for aggressively growing tumor types with poor prognosis. We improved the binding properties of a previously reported necrosis-avid contrast agent (NACA) and successfully detected spontaneous and therapy-induced tumor necrosis in mice using radioactivity and fluorescence imaging modalities. This NACA may pave the way to in vivo detection of tumor necrosis for early-stage determination of tumor aggressiveness and therapy efficacy. Abstract Purpose: To assess our improved NACA for the detection of tumor necrosis. Methods: We increased the blood circulation time of our NACA by adding an albumin-binding domain to the molecular structure. We tested the necrosis avidity on dead or alive cultured cells and performed SPECT and fluorescence imaging of both spontaneous and treatment-induced necrosis in murine breast cancer models. We simultaneously recorded [18F]FDG-PET and bioluminescence images for complementary detection of tumor viability. Results: We generated two albumin-binding IRDye800CW derivatives which were labeled with indium-111 with high radiochemical purity. Surprisingly, both albumin-binding NACAs had >10x higher in vitro binding towards dead cells. We selected [111In]3 for in vivo experiments which showed higher dead cell binding in vitro and in vivo stability. The doxorubicin-treated tumors showed increased [111In]3-uptake (1.74 ± 0.08%ID/g after saline treatment, 2.25 ± 0.16%ID/g after doxorubicin treatment, p = 0.044) and decreased [18F]FDG-uptake (3.02 ± 0.51%ID/g after saline treatment, 1.79 ± 0.11%ID/g after doxorubicin treatment, p = 0.040), indicating therapy efficacy. Moreover, we detected increased [111In]3-uptake and tumor necrosis in more rapidly growing EMT6 tumors. Conclusions: Our albumin-binding NACA based on IRDye800CW facilitates tumor-necrosis imaging for assessment of therapy efficacy and aggressiveness in solid tumors using both fluorescence and SPECT imaging.
Collapse
Affiliation(s)
- Marcus C. M. Stroet
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Erik de Blois
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
| | - Marion de Jong
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
| | - Yann Seimbille
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
| | - Laura Mezzanotte
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Clemens W. G. M. Löwik
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- CHUV Department of Oncology, University of Lausanne, CH-1066 Lausanne, Switzerland
- Correspondence: (C.W.G.M.L.); (K.M.P.)
| | - Kranthi M. Panth
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Correspondence: (C.W.G.M.L.); (K.M.P.)
| |
Collapse
|