1
|
Gong J, Li Y, Di H, Li J, Dong J, He C, Cao P, Cai H, He J, Wang Y. Combined PET and near-infrared fluorescence probe based on lapatinib targeting HER2 for in vivo tumor imaging. Bioorg Chem 2025; 161:108550. [PMID: 40367797 DOI: 10.1016/j.bioorg.2025.108550] [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: 03/07/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025]
Abstract
Combination of PET and optical imaging techniques holds significant potential for complete tumor resection to optimize patient outcomes. In this study, we combined two lapatinib-based molecular probes for detecting HER2-overexoression tumors through PET and near-infrared (NIR) fluorescence imaging. HER2 receptor had been confirmed to be overexpressed in many types of tumors and had become a specific target for molecular imaging. The radiolabeled probe [68Ga] Ga-NOTA-lapatinib demonstrated superior imaging performance in micro-PET scans, achieving clear differentiation of subcutaneous tumors from adjacent tissues. Complementarily, the NIR fluorescent probe LP-S exhibited prolonged tumor retention (>72 h) with distinct signal demarcation between malignant and normal tissues in ex vivo analyses. Fluorescence microscopy further validated specific intra-tumoral accumulation of the probe at cellular resolution. This synchronized dual-probe strategy, leveraging identical targeting moieties, established a robust platform for multimodality cancer imaging that bridges preoperative detection with intraoperative visualization.
Collapse
Affiliation(s)
- Jingyao Gong
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunlong Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Hui Di
- Zhangjiakou Food and Drug Inspection Center, Hebei, China
| | - Jiamin Li
- College of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Junming Dong
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Congjie He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huiming Cai
- Nanjing Nuoyuan Medical Devices Co. Ltd, Nanjing, China.
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Chen S, Xu H, Chen X, Shen Q, Chen X, Zhang M, Li Z, Zhang Z, Hao H, Yu W, Fan Y, Zhou L, Zhang N, Zhang J, Yang X, Shen C, Li X. First-in-human Study of a Dual-modality Prostate-specific Membrane Antigen-targeted Probe for Preoperative Positron Emission Tomography/Computed Tomography Imaging and Intraoperative Fluorescence Imaging in Prostate Cancer. Eur Urol 2025; 87:717-727. [PMID: 40204598 DOI: 10.1016/j.eururo.2025.03.010] [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: 01/09/2025] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND AND OBJECTIVE Accurately distinguishing between cancerous and noncancerous tissues during robot-assisted radical prostatectomy (RARP) is a challenge that can increase the risk of residual disease. This study aimed to evaluate the safety, optimal dose and accuracy of a dual-modality prostate-specific membrane antigen (PSMA)-targeted probe (68Ga-P3) for preoperative positron emission tomography (PET)/computed tomography (CT) imaging and intraoperative fluorescence imaging in prostate cancer. METHODS AND SURGICAL PROCEDURE Each participant received an intravenous chemical dose of 68Ga-P3 (10, 20, and 40 μg/kg), with radioactivity of 3.7 MBq/kg. PET/CT imaging was conducted 30, 60, and 120 min after injection to evaluate its biodistribution and dosimetry. RARP was performed at 24 ± 6 h after injection, in the sensitive mode of Firefly fluorescence imaging. KEY FINDINGS AND LIMITATIONS Between May 2024 and July 2024, a total of 16 patients were included; 68Ga-P3 was well tolerated without any adverse events related to 68Ga-P3 administration or fluorescence imaging. At 2 h after administration, the median tumor maximum standardized uptake value was 5.3 (4.1-8.1). The sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of 68Ga-P3 PET/CT for tumor localization were 79.1%, 90.4%, 81.5%, and 89.0%, respectively. The overall NPV, PPV, and accuracy of intraoperative fluorescence imaging were 100%, 43.8%, and 90.9%, respectively. Of overall false-positive sites, 88.9% (8/9) were confirmed as tumor adjacent to the surgical margin. A dose of 40 μg/kg resulted in the highest accuracy of 92.3%. CONCLUSIONS AND CLINICAL IMPLICATIONS In PSMA-targeted PET imaging and fluorescence-guided surgery, 68Ga-P3 is safe and feasible for use, offering a novel tool for the surgical management of prostate cancer.
Collapse
Affiliation(s)
- Silu Chen
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Hongchuang Xu
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng District, Beijing, China
| | - Xueqi Chen
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng District, Beijing, China
| | - Qi Shen
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Xu Chen
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Meng Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Zhihua Li
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Zhongyuan Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Han Hao
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Wei Yu
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Yan Fan
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng District, Beijing, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China
| | - Ning Zhang
- Translational Cancer Research Center, Peking University First Hospital, Xicheng District, Beijing, China
| | - Jianhua Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng District, Beijing, China.
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, Xicheng District, Beijing, China; Department of Nuclear Medicine, Peking University People's Hospital, Xicheng District, Beijing, China.
| | - Cheng Shen
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China.
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Xicheng District, Beijing, China.
| |
Collapse
|
3
|
Li Z, Peng T, Yang M, Qiu Y, Ye P, Wang X, Jin H. Dual Functionality of [ 64Cu]Cu-NOTA-San A-Cy7 for Diagnostic Imaging and Surgical Guidance in Hsp90α-Positive Tumors. Mol Pharm 2025. [PMID: 40343454 DOI: 10.1021/acs.molpharmaceut.5c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Intraoperative fluorescence navigation in esophageal cancer enables the clinical translation of fluorescence imaging. Heat shock protein 90 alpha (Hsp90α) plays a vital role in the progression of malignant disease, and elevated Hsp90α expression has been reported in esophageal cancer. The aim of this study was to develop a dual-modality probe, [64Cu]Cu-NOTA-San A-Cy7, for imaging Hsp90α expression in vivo via both positron emission tomography (PET) and fluorescence imaging in esophageal cancer. In this study, the Hsp90α-targeting cyclopeptide Sansalvamide A (San A) was chemically modified with a Cy7 dye and NOTA chelator simultaneously. Experimental assays confirmed that NOTA-San A-Cy7 has a favorable affinity for Hsp90α-positive EC109 cells, with a dissociation constant (Kd) of 1.08 ± 0.19 μM. The probe [64Cu]Cu-NOTA-San A-Cy7 was successfully synthesized with 64CuCl2, achieving a high radiochemical purity of over 95%. Furthermore, the probe demonstrated excellent stability in both saline and serum solutions. The probe was subsequently evaluated in a Hsp90α-positive EC109 tumor-bearing model via PET imaging, which confirmed that Hsp90α-specific uptake was significantly reduced by the co-administration of an excess blocking agent. Biodistribution studies revealed that at 24 hours post-injection, the tumor uptake of the probe was 1.35 ± 0.29%ID/g in the nonblocking group and significantly decreased to 0.73 ± 0.15%ID/g in the blocking group (p < 0.05). Concurrent with the PET experiment, fluorescence imaging was conducted, revealing substantial tumor uptake in the EC109 model. As a proof of concept, imaging-guided surgery utilizing the fluorescent component of this probe was performed. This approach demonstrated the potential for providing surgical guidance in mice positive for Hsp90α, highlighting the dual functionality of the probe for both diagnostic imaging and intraoperative navigation. In summary, our findings unequivocally demonstrate that the dual-modality probe [64Cu]Cu-NOTA-San A-Cy7 holds significant promise as an agent for imaging Hsp90α-positive tumors in vivo, offering a valuable tool for the detection and potential management of such tumors.
Collapse
Affiliation(s)
- Zhijun Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
- Department of Urology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan Province 421001, China
| | - Tukang Peng
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Min Yang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yifan Qiu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Peizhen Ye
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaohui Wang
- Department of Nuclear Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| |
Collapse
|
4
|
Quarta L, Cannoletta D, Pellegrino F, Barletta F, Scuderi S, Mazzone E, Stabile A, Montorsi F, Gandaglia G, Briganti A. The Role of Robot-Assisted, Imaging-Guided Surgery in Prostate Cancer Patients. Cancers (Basel) 2025; 17:1401. [PMID: 40361328 PMCID: PMC12070902 DOI: 10.3390/cancers17091401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Emerging imaging-guided technologies, such as prostate-specific membrane antigen radioguided surgery (PSMA-RGS) and augmented reality (AR), could enhance the precision and efficacy of robot-assisted prostate cancer (PCa) surgical approaches, maximizing the surgeons' ability to remove all cancer sites and thus patients' outcomes. Sentinel node biopsy (SNB) represents an imaging-guided technique that could enhance nodal staging accuracy by leveraging lymphatic mapping with tracers. PSMA-RGS uses radiolabeled tracers with the aim to improve intraoperative lymph node metastases (LNMs) detection. Several studies demonstrated its feasibility and safety, with promising accuracy in nodal staging during robot-assisted radical prostatectomy (RARP) and in recurrence setting during salvage lymph node dissection (sLND) in patients who experience biochemical recurrence (BCR) after primary treatment and have positive PSMA positron emission tomography (PET). Near-infrared PSMA tracers, such as OTL78 and IS-002, have shown potential in intraoperative fluorescence-guided surgery, improving positive surgical margins (PSMs) and LNMs identification. Finally, augmented reality (AR), which integrates preoperative imaging (e.g., multiparametric magnetic resonance imaging [mpMRI] of the prostate and computed tomography [CT]) onto the surgical field, can provide a real-time visualization of anatomical structures through the creation of three-dimensional (3D) models. These technologies may assist surgeons during intraoperative procedures, thus optimizing the balance between oncological control and functional outcomes. However, challenges remain in standardizing these tools and assessing their impact on long-term PCa control. Overall, these advancements represent a paradigm shift toward personalized and precise surgical approaches, emphasizing the integration of innovative strategies to improve outcomes of PCa patients.
Collapse
Affiliation(s)
- Leonardo Quarta
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Donato Cannoletta
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Pellegrino
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Barletta
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Simone Scuderi
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elio Mazzone
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Armando Stabile
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Montorsi
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Giorgio Gandaglia
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alberto Briganti
- Unit of Urology, Division of Oncology, Gianfranco Soldera Prostate Cancer Lab, URI, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
5
|
Sun JX, Xia QD, Xu JZ, An Y, Ma SY, Xu JY, Xiang JC, Liu CQ, Xu MY, Zhang SH, Luan Y, Tang K, Wang SG. A novel prostate cancer-specific fluorescent probe based on extracellular vesicles targeting STEAP1 applied in fluorescence guided surgery. J Control Release 2025; 380:199-218. [PMID: 39894263 DOI: 10.1016/j.jconrel.2025.01.079] [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: 11/13/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Radical prostatectomy with pelvic lymph node dissection is the best treatment for intermediate- to high-risk localized prostate cancer (PCa). However, conventional white light surgery has difficulties in identifying tumor boundary and micrometastases intraoperatively. Fluorescence guided surgery (FGS) can solve the above difficulties, but lacks tumor-specific near-infrared fluorescent (NIRF) probes in PCa. STEAP1 was an ideal target in PCa treatment and imaging. Here, we constructed a PCa specific fluorescent probe based on extracellular vesicles targeting STEAP1 (AS-EVs) loaded with NIRF dye S0456 and evaluated its preclinical profiles. In vitro and in vivo studies both showed S0456@AS-EVs was safe and showed strong targeting ability to PCa in various mice xenograft models. S0456@AS-EVs could clear rapidly from blood (half-time of 4.29 h) and retain in the STEAP1 positive tumor tissues for more than 72 h with the highest tumor background ratio (TBR) of 3:1, which was superior to ICG, free S0456, ICG@Ctrl-EVs and S0456@Ctrl-EVs (p < 0.01). Finally, S0456@AS-EVs was applied in FGS on intramuscular model, and the tumors were resected under white light and fluorescence respectively. Compared with white light surgery, mice undergoing FGS had lower positive margin rate and better postoperative survival (p = 0.0342).
Collapse
Affiliation(s)
- Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jing-Yu Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jia-Cheng Xiang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, USA
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yang Luan
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
6
|
Minges P, Eder M, Eder AC. Dual-Labeled Small Peptides in Cancer Imaging and Fluorescence-Guided Surgery: Progress and Future Perspectives. Pharmaceuticals (Basel) 2025; 18:143. [PMID: 40005958 PMCID: PMC11858487 DOI: 10.3390/ph18020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Dual-labeled compounds that combine radiolabeling and fluorescence labeling represent a significant advancement in precision oncology. Their clinical implementation enhances patient care and outcomes by leveraging the high sensitivity of radioimaging for tumor detection and taking advantage of fluorescence-based optical visualization for surgical guidance. Non-invasive radioimaging facilitates immediate identification of both primary tumors and metastases, while fluorescence imaging assists in decision-making during surgery by offering a spatial distinction between malignant and non-malignant tissue. These advancements hold promise for enhancing patient outcomes and personalization of cancer treatment. The development of dual-labeled molecular probes targeting various cancer biomarkers is crucial in addressing the heterogeneity inherent in cancer pathology and recent studies had already demonstrated the impact of dual-labeled compounds in surgical decision-making (NCT03699332, NCT03407781). This review focuses on the development and application of small dual-labeled peptides in the imaging and treatment of various cancer types. It summarizes the biomarkers targeted to date, tracing their development from initial discovery to the latest advancements in peptidomimetics. Through comprehensive analysis of recent preclinical and clinical studies, the review demonstrates the potential of these dual-labeled peptides to improve tumor detection, localization, and resection. Additionally, it highlights the evolving landscape of dual-modality imaging, emphasizing its critical role in advancing personalized and effective cancer therapy. This synthesis of current research underscores the promise of dual-labeled peptides in enhancing diagnostic accuracy and therapeutic outcomes in oncology.
Collapse
Affiliation(s)
- Paul Minges
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (P.M.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (P.M.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (P.M.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
7
|
Zhang X, Ma L, Cai K, Guo X, Zhang G, Dong J, Zheng Y, Su X, Tao T, Li X, Yuan Y. PSMA-Targeted Intracellular Self-Assembled Probe for Enhanced PET Imaging. Bioconjug Chem 2025; 36:20-24. [PMID: 39810600 DOI: 10.1021/acs.bioconjchem.4c00572] [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: 01/16/2025]
Abstract
Positron-emission tomography (PET) offers high sensitivity for cancer diagnosis. However, small-molecule-based probes often exhibit insufficient accumulation in tumor sites, while nanoparticle-based agents typically have limited delivery efficiency. To address this challenge, this study proposes a novel PET imaging probe, 68Ga-CBT-PSMA, designed for prostate cancer. This probe integrates an intracellular self-assembly strategy to enhance PET imaging signals and significantly improve the signal-to-noise ratio. The glutamate-urea-based prostate-specific membrane antigen (PSMA)-targeting motif enables specific recognition of prostate cancer cells and enhances cellular uptake; then the self-assembly process induced by glutathione reduction effectively accumulates the probe within tumor cells, thereby amplifying PET imaging signals. This approach not only enhances signal intensity and resolution but also facilitates precise cancer localization and diagnosis, offering new avenues for advancing cancer diagnostic techniques.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Li Ma
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Ke Cai
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiangyuan Guo
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Guangtao Zhang
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Jiajing Dong
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Yifan Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiaoyu Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Tao Tao
- Department of Urology, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, China
| | - Xiaohu Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yue Yuan
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
8
|
Kerpa S, Holzapfel M, Staufer T, Kuhrwahl R, Mutas M, Werner S, Schulze VR, Nakielski P, Feliu N, Oetjen E, Haak J, Ziegler F, Buchin R, Han J, Parak WJ, Grüner F, Maison W. Iodinated PSMA Ligands as XFI Tracers for Targeted Cell Imaging and Characterization of Nanoparticles. Int J Mol Sci 2024; 25:11880. [PMID: 39595950 PMCID: PMC11594147 DOI: 10.3390/ijms252211880] [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: 03/25/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men worldwide. Despite this, current diagnostic tools are still not satisfactory, lacking sensitivity for early-stage or single-cell diagnosis. This study describes the development of small-molecule tracers for the well-known tumor marker prostate-specific membrane antigen (PSMA). These tracers contain a urea motif for PSMA-targeting and iodinated aromatic moieties to allow detection via X-ray fluorescence imaging (XFI). Tracers with a triiodobenzoyl moiety allowed the specific targeting and successful imaging of PSMA+ cell lines with XFI. The XFI-measured uptake of 7.88 × 10-18 mol iodine (I) per cell is consistent with the uptake of known PSMA tracers measured by other techniques such as inductively coupled plasma mass spectrometry (ICP-MS). This is the first successful application of XFI to tumor cell targeting with a small-molecule tracer. In addition, iodinated tracers were used for the characterization of quantum dots (QDs) conjugated to PSMA-targeting urea motifs. The resulting targeted QD conjugates were shown to selectively bind PSMA+ cell lines via confocal microscopy. The immobilized iodinated targeting vectors allowed the determination of the tracer/QD ratio via XFI and ICP-MS. This ratio is a key property of targeted particles and difficult to measure by other techniques.
Collapse
Affiliation(s)
- Svenja Kerpa
- Department of Chemistry, Institute of Pharmacy, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany;
| | - Malte Holzapfel
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Theresa Staufer
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Robert Kuhrwahl
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Marina Mutas
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Verena R. Schulze
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Pascal Nakielski
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Neus Feliu
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Elke Oetjen
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany;
| | - Jannis Haak
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Florian Ziegler
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Rasmus Buchin
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Jili Han
- Department of Physics, Universität Hamburg and Center for Hybrid Nanostructures (CHyN), Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (W.J.P.)
| | - Wolfgang J. Parak
- Department of Physics, Universität Hamburg and Center for Hybrid Nanostructures (CHyN), Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (W.J.P.)
| | - Florian Grüner
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Wolfgang Maison
- Department of Chemistry, Institute of Pharmacy, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany;
| |
Collapse
|
9
|
Dinneen E, Almeida-Magana R, Al-Hammouri T, Fernandes I, Mayor N, Mendes L, Winkler M, Silvanto A, Haider A, Freeman A, Shaw G. Intraoperative margin assessment during radical prostatectomy: is microscopy frozen in time or ready for digital defrost? Histopathology 2024; 85:716-726. [PMID: 39104212 DOI: 10.1111/his.15290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Intraoperative frozen section (IFS) is used with the intention to improve functional and oncological outcomes for patients undergoing radical prostatectomy (RP). High resource requirements of IFS techniques such as NeuroSAFE may preclude widespread adoption, even if there are benefits to patients. Recent advances in fresh-tissue microscopic digital imaging technologies may offer an attractive alternative, and there is a growing body of evidence regarding these technologies. In this narrative review, we discuss some of the familiar limitations of IFS and compare these to the attractive counterpoints of modern digital imaging technologies such as the speed and ease of image generation, the locality of equipment within (or near) the operating room, the ability to maintain tissue integrity, and digital transfer of images. Confocal laser microscopy (CLM) is the modality most frequently reported in the literature for margin assessment during RP. We discuss several imitations and obstacles to widespread dissemination of digital imaging technologies. Among these, we consider how the 'en-face' margin perspective will challenge urologists and pathologists to understand afresh the meaning of positive margin significance. As a part of this, discussions on how to describe, categorize, react to, and evaluate these technologies are needed to improve patient outcomes. Limitations of this review include its narrative structure and that the evidence base in this field is relatively immature but developing at pace.
Collapse
Affiliation(s)
- Eoin Dinneen
- Division of Surgery & Interventional Science, University College London, London, UK
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| | - Ricardo Almeida-Magana
- Division of Surgery & Interventional Science, University College London, London, UK
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| | - Tarek Al-Hammouri
- Division of Surgery & Interventional Science, University College London, London, UK
- Centre for Medical Imaging, University College London, London, UK
| | - Iona Fernandes
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| | - Nikhil Mayor
- Department of Urology, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
- Imperial Prostate, Division of Surgery, Imperial College London, London, UK
| | - Larissa Mendes
- Department of Histopathology, University College Hospital London, London, UK
| | - Mathias Winkler
- Department of Urology, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
- Imperial Prostate, Division of Surgery, Imperial College London, London, UK
| | - Anna Silvanto
- Department of Histopathology, University College Hospital London, London, UK
| | - Aiman Haider
- Department of Histopathology, University College Hospital London, London, UK
| | - Alex Freeman
- Department of Histopathology, University College Hospital London, London, UK
| | - Greg Shaw
- Division of Surgery & Interventional Science, University College London, London, UK
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| |
Collapse
|
10
|
Kong S, Peng Y, Liu Q, Xie Q, Qiu L, Lin J, Xie M. Preclinical Evaluation of a PSMA Aptamer-Based Bifunctional PET and Fluorescent Probe. Bioconjug Chem 2024; 35:1352-1362. [PMID: 39187748 DOI: 10.1021/acs.bioconjchem.4c00292] [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: 08/28/2024]
Abstract
Prostate cancer is the most prevalent malignant tumor affecting male individuals worldwide. The accurate early detection of prostate cancer is crucial to preventing unnecessary diagnosis and subsequent excessive treatment. Prostate-specific membrane antigen (PSMA) has emerged as a promising biomarker for the diagnosis of prostate cancer. In this study, a dual-modality imaging probe utilizing aptamer technology was developed for positron emission tomography/near-infrared fluorescence (PET/NIRF) imaging, and the specificity and sensitivity of the probe toward PSMA were evaluated both in vitro and in vivo. The probe precursor NOTA-PSMA-Cy5 was synthesized via automated solid-phase oligonucleotide synthesis. Subsequently, the PET/NIRF dual-modality probe [68Ga]Ga-NOTA-PSMA-Cy5 was successfully prepared and exhibited favorable fluorescence properties and stability in vitro. The binding specificity of [68Ga]Ga-NOTA-PSMA-Cy5 to PSMA was assessed through flow cytometry, fluorescence imaging, and cellular uptake experiments in LNCaP cells and PC-3 cells. In vivo PET/NIRF imaging studies demonstrated the sensitive and specific binding of [68Ga]Ga-NOTA-PSMA-Cy5 to PSMA. Overall, the PET/NIRF dual-modality probe [68Ga]Ga-NOTA-PSMA-Cy5 shows promise for the diagnosis of prostate cancer and for the fluorescence-guided identification of PSMA-positive cancer lesions during surgical procedures.
Collapse
Affiliation(s)
- Sudong Kong
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
- Suzhou Biosyntech Co., Ltd., Suzhou 215300, China
| | - Ying Peng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Qingzhu Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Quan Xie
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Ling Qiu
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Jianguo Lin
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Minhao Xie
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
11
|
Lewis MR, Schaedler AW, Ho KV, Golzy M, Mathur A, Pun M, Gallazzi F, Watkinson LD, Carmack TL, Sikligar K, Anderson CJ, Smith CJ. Evaluation of a bimodal, matched pair theranostic agent targeting prostate-specific membrane antigen. Nucl Med Biol 2024; 136-137:108938. [PMID: 39032262 DOI: 10.1016/j.nucmedbio.2024.108938] [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: 03/05/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Prostate cancer affects 1 in 6 men, and it is the second‑leading cause of cancer-related death in American men. Surgery is one of the main treatment modalities for prostate cancer, but it often results in incomplete resection margins or complete resection that leads to nerve damage and undesirable side effects. In the present work, we have developed a new bimodal tracer, NODAGA-sCy7.5 PSMAi (prostate-specific membrane antigen inhibitor), labeled with the true matched theranostic pair 64Cu/67Cu and a near-infrared fluorescent dye. This agent could potentially be used for concomitant PET imaging, optical surgical navigation, and targeted radiopharmaceutical therapy. METHODS A prostate-specific membrane antigen (PSMA)-targeting urea derivative was conjugated to NODAGA for copper radiolabeling and to the near-infrared fluorophore sulfo-Cy7.5 (sCy7.5). Binding studies were performed in PSMA-positive PC-3 PIP cells, as well as uptake and internalization assays in PC-3 PIP cells and PSMA-negative PC-3 wild type cells. Biodistribution studies of the 64Cu-labeled compound were performed in PC-3 PIP- and PC-3 tumor-bearing mice, and 67Cu biodistributions of the agent were obtained in PC-3 PIP tumor-carrying mice. PET imaging and fluorescence imaging were also performed, using the same molar doses, in the two mouse models. RESULTS The PSMA conjugate bound with high affinity to PSMA-positive prostate cancer cells, as opposed to cells that were PSMA-negative. Uptake and internalization were rapid and PSMA-mediated in PC-3 PIP cells, while only minimal non-specific uptake was observed in PC-3 cells. Biodistribution studies showed specific uptake in PC-3 PIP tumors, while accumulation in PC-3 tumor-bearing mice was low. Furthermore, tumor uptake of the 67Cu-labeled agent in the PC-3 PIP model was statistically equivalent to that of 64Cu. PET and fluorescence imaging at 0.5 nmol per mouse also demonstrated that PC-3 PIP tumors could be clearly detected, while PC-3 tumors showed no tumor accumulation. CONCLUSIONS NODAGA-sCy7.5-PSMAi was specific and selective in detecting PSMA-positive, as opposed to PSMA-negative, tumors in mouse models of prostate cancer. This bioconjugate could potentially be used for PET staging with 64Cu, targeted radiopharmaceutical therapy with 67Cu, and/or image-guided surgery with sCy7.5.
Collapse
Affiliation(s)
- Michael R Lewis
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, United States of America; Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, United States of America.
| | - Alexander W Schaedler
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, United States of America; Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America
| | - Khanh-Van Ho
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Department of Chemistry, University of Missouri, Columbia, MO, United States of America
| | - Mojgan Golzy
- Biostatistics Unit, University of Missouri, Columbia, MO, United States of America
| | - Anupam Mathur
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Research Reactor Center, Columbia, MO, United States of America
| | - Michael Pun
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Department of Chemistry, University of Missouri, Columbia, MO, United States of America
| | - Fabio Gallazzi
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Molecular Interactions Core, University of Missouri, Columbia, MO, United States of America
| | - Lisa D Watkinson
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Research Reactor Center, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America
| | - Terry L Carmack
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Research Reactor Center, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America
| | - Kanishka Sikligar
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Department of Chemistry, University of Missouri, Columbia, MO, United States of America
| | - Carolyn J Anderson
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, United States of America; Department of Chemistry, University of Missouri, Columbia, MO, United States of America; Department of Radiology, University of Missouri, Columbia, MO, United States of America
| | - Charles J Smith
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Department of Radiology, University of Missouri, Columbia, MO, United States of America
| |
Collapse
|
12
|
Buckle T, Rietbergen DDD, de Wit-van der Veen L, Schottelius M. Lessons learned in application driven imaging agent design for image-guided surgery. Eur J Nucl Med Mol Imaging 2024; 51:3040-3054. [PMID: 38900308 PMCID: PMC11300579 DOI: 10.1007/s00259-024-06791-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
To meet the growing demand for intraoperative molecular imaging, the development of compatible imaging agents plays a crucial role. Given the unique requirements of surgical applications compared to diagnostics and therapy, maximizing translational potential necessitates distinctive imaging agent designs. For effective surgical guidance, exogenous signatures are essential and are achievable through a diverse range of imaging labels such as (radio)isotopes, fluorescent dyes, or combinations thereof. To achieve optimal in vivo utility a balanced molecular design of the tracer as a whole is required, which ensures a harmonious effect of the imaging label with the affinity and specificity (e.g., pharmacokinetics) of a pharmacophore/targeting moiety. This review outlines common design strategies and the effects of refinements in the molecular imaging agent design on the agent's pharmacological profile. This includes the optimization of affinity, pharmacokinetics (including serum binding and target mediated background), biological clearance route, the achievable signal intensity, and the effect of dosing hereon.
Collapse
Affiliation(s)
- Tessa Buckle
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda de Wit-van der Veen
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Rue du Bugnon 25A, Agora, Lausanne, CH-1011, Switzerland.
- Agora, pôle de recherche sur le cancer, Lausanne, Switzerland.
| |
Collapse
|
13
|
Fu H, Lou K, He H, Wang Y, Mi Y, Li W, Chen L, Zhang Y, Yu C. A novel PSMA targeted dual-function near-infrared fluorescence and PET probe for the image-guided surgery and detection of prostate cancer. Eur J Nucl Med Mol Imaging 2024; 51:2998-3008. [PMID: 37914976 DOI: 10.1007/s00259-023-06492-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is a promising diagnostic biomarker for prostate cancer (PCa). NYM016, a novel small-molecule PSMA-targeted fluorescence probe for the surgical navigation of PCa, was designed in this work. Furthermore, the potential of the PET agent [68Ga]Ga-NYM016 for the radionuclide imaging of PCa was evaluated. METHODS NYM016 was designed with the near-infrared fluorescent group Cyanine 7 (Cy7) and the chelating group NOTA. The radioactive probe [68Ga]Ga-NYM016 was designed and synthesized on the basis of NYM016. The abovementioned probes were assessed in PSMA-positive xenograft-bearing models and patients diagnosed with PCa. RESULTS NYM016 obviously aggregated in the tumor site of the mouse model, and its fluorescence intensity was stable within 24 h. NYM016 was well-tolerated, and no adverse events were found in the clinical study. Moreover, it was also observed in the excised lesions from the patient with PCa, and its fluorescence aggregated at the same site where PSMA was highly expressed. In addition, the PSMA xenograft demonstrated intense [68Ga]Ga-NYM016 uptake at 2.5 min after injection. At 3 h after injection, [68Ga]Ga-NYM016 uptake by the PSMA xenograft gradually increased to 6.40 ± 0.19%ID/g, which was higher that by the blocked and negative groups (2.28 ± 0.07%ID/g, P < 0.05; 2.28 ± 0.22%ID/g, P < 0.05). In the clinical study, [68Ga]Ga-NYM016 was well-tolerated and no adverse events were observed. Substantial accumulation was observed in primary and metastatic lesions in a patient with recurrence with the maximum standardized uptake value of 18.93. Meanwhile, negative [68Ga]Ga-NYM016 uptake was observed at the prostate site of a patient with prostatitis. CONCLUSION The novel fluorescence probe NYM016 and the radioactive tracer [68Ga]Ga-NYM016 are promising candidates for the surgical navigation and radionuclide imaging of PCa, respectively. TRIAL REGISTRATION The clinical evaluation of this study was registered at Clinicaltrial.gov (NCT05623878) on 21 Dec, 2022.
Collapse
Affiliation(s)
- Haitian Fu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Kequan Lou
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Huihui He
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Yanjuan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Yuanyuan Mi
- Department of Urological Surgery, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Wenjin Li
- Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, 214000, China
| | - Liping Chen
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Yu Zhang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214000, China.
- Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, 214000, China.
| |
Collapse
|
14
|
Zhang X, Huang J, Gong F, Cai Z, Liu Y, Tang G, Hu K. Synthesis and preclinical evaluation of a novel PET/fluorescence dual-modality probe targeting fibroblast activation protein. Bioorg Chem 2024; 146:107275. [PMID: 38493637 DOI: 10.1016/j.bioorg.2024.107275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Early diagnosis and precise surgical intervention are crucial for cancer patients. We aimed to develop a novel positron emission tomography (PET)/fluorescence dual-modality probe for preoperative diagnosis, intraoperative guidance, and postoperative monitoring of fibroblast activation protein (FAP)-positive tumors. FAPI-FAM was synthesized and labeled with gallium-68. [68Ga]Ga-FAPI-FAM showed favorable in vivo and in vitro characteristics, specific binding affinity, and excellent tumor accumulation in FAP-positive cells and mice xenografts. Excellent tumor-to-background contrast was found owing to high tumor uptake, prolonged retention, and rapid renal clearance of [68Ga]Ga-FAPI-FAM. Moreover, a specific fluorescence signal was detected in FAP-positive tumors during ex vivo fluorescence imaging, demonstrating the feasibility of whole-body tumor detection and intraoperative tumor delineation.
Collapse
Affiliation(s)
- Xiaojun Zhang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Jiawen Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Fengping Gong
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Zhikai Cai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Yang Liu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Ganghua Tang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| | - Kongzhen Hu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| |
Collapse
|
15
|
Lengacher R, Martin KE, Śmiłowicz D, Esseln H, Lotlikar P, Grichine A, Maury O, Boros E. Targeted, Molecular Europium (III) Probes Enable Luminescence-Guided Surgery and 1 Photon Post-Surgical Luminescence Microscopy of Solid Tumors. J Am Chem Soc 2023; 145:24358-24366. [PMID: 37869897 PMCID: PMC10670433 DOI: 10.1021/jacs.3c09444] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Discrete luminescent lanthanide complexes represent a potential alternative to organic chromophores due to their tunability of optical properties, insensitivity to photobleaching, and large pseudo-Stokes shifts. Previously, we demonstrated that the lack of depth penetration of UV excitation required to sensitize discrete terbium and europium complexes can be overcome using Cherenkov radiation emitted by clinically employed radioisotopes in situ. Here, we show that the second-generation europium complexes [Eu(III)(pcta-PEPA2)] and [Eu(III)(tacn-pic-PEPA2)] (Φ = 57% and 76%, respectively) lower the limit of detection (LoD) to 1 nmol in the presence of 10 μCi of Cherenkov emitting isotopes, 18F and 68Ga. Bifunctionalization provides access to cysteine-linked peptide conjugates with comparable brightness and LoD. The conjugate, [Eu(tacn-(pic-PSMA)-PEPA2)], displays high binding affinity to prostate-specific membrane antigen (PSMA)-expressing PC-3 prostate cancer cells in vitro and can be visualized in the membrane-bound state using confocal microscopy. Biodistribution studies with the [86Y][Y(III)(tacn-(pic-PSMA)-PEPA2)] analogue in a mouse xenograft model were employed to study pharmacokinetics. Systemic administration of the targeted Cherenkov emitter, [68Ga][Ga(III)(PSMA-617)], followed by intratumoral injection or topical application of 20 or 10 nmol [Eu(III)(tacn-(pic-PSMA)-PEPA2)], respectively, in live mice resulted in statistically significant signal enhancement using conventional small animal imaging (620 nm bandpass filter). Optical imaging informed successful tumor resection. Ex vivo imaging of the fixed tumor tissue with 1 and 2 photon excitation further reveals the accumulation of the administered Eu(III) complex in target tissues. This work represents a significant step toward the application of luminescent lanthanide complexes for optical imaging in a clinical setting.
Collapse
Affiliation(s)
- Raphael Lengacher
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Kirsten E Martin
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Dariusz Śmiłowicz
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Helena Esseln
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Piyusha Lotlikar
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Alexei Grichine
- Institute for Advanced Biosciences, Université Grenoble Alpes, Inserm U1209, CNRS, UMR 5309, Site Santé, Allée des Alpes, 38700 La Tronche, France
| | - Olivier Maury
- Université Lyon, ENS de Lyon, CNRS, Laboratoire de Chimie UMR 5182, F-69342 Lyon, France
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| |
Collapse
|
16
|
Sullivan TE, Hernandez Vargas S, Ghosh SC, AghaAmiri S, Ikoma N, Azhdarinia A. A translational blueprint for developing intraoperative imaging agents via radiopharmaceutical-guided drug design. Curr Opin Chem Biol 2023; 76:102376. [PMID: 37572489 DOI: 10.1016/j.cbpa.2023.102376] [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: 02/09/2023] [Revised: 06/22/2023] [Accepted: 07/07/2023] [Indexed: 08/14/2023]
Abstract
Cancer imaging is a rapidly evolving field due to the discovery of novel molecular targets and the availability of corresponding techniques to detect them with high precision, accuracy, and sensitivity. Nuclear medicine is the most widely used molecular imaging modality and has a growing toolkit of clinically used radiopharmaceuticals that enable whole-body tumor visualization, staging, and treatment monitoring for a variety of tumors in a non-invasive manner. The need for similar imaging capabilities in the operating room has led to the emergence of fluorescence-guided surgery (FGS) as a powerful technique that gives surgeons unprecedented ability to distinguish tumors from healthy tissues. While a variety of strategies have been used to develop contrast agents for FGS, the use of radiopharmaceuticals as models brings exceptional translational potential and has increasingly been explored. Here, we review strategies used to convert clinically used radiopharmaceuticals into fluorescent and multimodal counterparts. Unique preclinical and clinical capabilities stemming from radiopharmaceutical-based agent design are also discussed to illustrate the advantages of this approach.
Collapse
Affiliation(s)
- Teresa E Sullivan
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Sukhen C Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.
| |
Collapse
|
17
|
Zyk NY, Garanina AS, Plotnikova EA, Ber AP, Nimenko EA, Dashkova NS, Uspenskaia AA, Shafikov RR, Skvortsov DA, Petrov SA, Pankratov AA, Zyk NV, Majouga AG, Beloglazkina EK, Machulkin AE. Synthesis of Prostate-Specific Membrane Antigen-Targeted Bimodal Conjugates of Cytotoxic Agents and Antiandrogens and Their Comparative Assessment with Monoconjugates. Int J Mol Sci 2023; 24:11327. [PMID: 37511087 PMCID: PMC10380083 DOI: 10.3390/ijms241411327] [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: 05/30/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer is the second most common cancer among men. We designed and synthesized new ligands targeting prostate-specific membrane antigen and suitable for bimodal conjugates with diagnostic and therapeutic agents. In vitro studies of the affinity of the synthesized compounds to the protein target have been carried out. Based on these ligands, a series of bimodal conjugates with a combination of different mitosis inhibitors and antiandrogenic drugs were synthesized. The cytotoxicity of the compounds obtained in vitro was investigated on three different cell lines. The efficacy of the two obtained conjugates was evaluated in vivo in xenograft models of prostate cancer. These compounds have been shown to be highly effective in inhibiting the growth of PSMA-expressing tumors.
Collapse
Affiliation(s)
- Nikolai Y Zyk
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Anastasiia S Garanina
- Laboratory of Biomedical Nanomaterials, National University of Science and Technology MISIS, 4 Leninskiy pr, Moscow 119049, Russia
| | - Ekaterina A Plotnikova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 2 Botkinskiy proezd, 3, Moscow 125284, Russia
| | - Anton P Ber
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Ekaterina A Nimenko
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Natalia S Dashkova
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Anastasiia A Uspenskaia
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Radik R Shafikov
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, GSP-7, Ulitsa Miklukho-Maklaya, 16/10, Moscow 117997, Russia
| | - Dmitry A Skvortsov
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Stanislav A Petrov
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Andrey A Pankratov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 2 Botkinskiy proezd, 3, Moscow 125284, Russia
| | - Nikolai V Zyk
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Alexander G Majouga
- Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russia
| | - Elena K Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| | - Aleksei E Machulkin
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russia
| |
Collapse
|
18
|
Uspenskaia AA, Krasnikov PA, Majouga AG, Beloglazkina EK, Machulkin AE. Fluorescent Conjugates Based on Prostate-Specific Membrane Antigen Ligands as an Effective Visualization Tool for Prostate Cancer. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:953-967. [PMID: 37751866 DOI: 10.1134/s0006297923070088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 09/28/2023]
Abstract
Fluorescent dyes are widely used in histological studies and in intraoperative imaging, including surgical treatment of prostate cancer (PC), which is one of the most common types of cancerous tumors among men today. Targeted delivery of fluorescent conjugates greatly improves diagnostic efficiency and allows for timely correct diagnosis. In the case of PC, the protein marker is a prostate-specific membrane antigen (PSMA). To date, a large number of diagnostic conjugates targeting PSMA have been created based on modified urea. The review focuses on the conjugates selectively binding to PSMA and answers the following questions: What fluorescent dyes are already in use in the field of PC diagnosis? What factors influence the structure-activity ratio of the final molecule? What features should be considered when selecting a fluorescent tag to create new diagnostic conjugates? And what could be suggested to further development in this field at the present time?
Collapse
Affiliation(s)
| | - Pavel A Krasnikov
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexander G Majouga
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
- National University of Science and Technology "MISiS", Moscow, 119049, Russia
- Dmitry Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russia
| | | | - Aleksei E Machulkin
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
- RUDN University, Moscow, 117198, Russia
| |
Collapse
|
19
|
d'Orchymont F, Holland JP. Asymmetric rotaxanes as dual-modality supramolecular imaging agents for targeting cancer biomarkers. Commun Chem 2023; 6:107. [PMID: 37264077 DOI: 10.1038/s42004-023-00906-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/17/2023] [Indexed: 06/03/2023] Open
Abstract
Dual-modality imaging agents featuring both a radioactive complex for positron emission tomography (PET) and a fluorophore for optical fluorescence imaging (OFI) are crucial tools for reinforcing clinical diagnosis and intraoperative surgeries. We report the synthesis and characterisation of bimodal mechanically interlocked rotaxane-based imaging agents, constructed via the cucurbit[6]uril CB[6]-mediated alkyne-azide 'click' reaction. Two synthetic routes involving four- or six-component reactions are developed to access asymmetric rotaxanes. Furthermore, by using this rapid and versatile approach, a peptide-based rotaxane targeted toward the clinical prostate cancer biomarker, prostate-specific membrane antigen (PSMA), and bearing a 68Ga-radiometal ion complex for positron emission tomography and fluorescein as an optically active imaging agent, was synthesised. The chemical and radiochemical stability, and the cellular uptake profile of the radiolabelled and fluorescent rotaxane was evaluated in vitro where the experimental data demonstrate the viability of using an asymmetric rotaxane platform to produce dual-modality imaging agents that specifically target prostate cancer cells.
Collapse
Affiliation(s)
- Faustine d'Orchymont
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Jason P Holland
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
20
|
Stibbe JA, de Barros HA, Linders DGJ, Bhairosingh SS, Bekers EM, van Leeuwen PJ, Low PS, Kularatne SA, Vahrmeijer AL, Burggraaf J, van der Poel HG. First-in-patient study of OTL78 for intraoperative fluorescence imaging of prostate-specific membrane antigen-positive prostate cancer: a single-arm, phase 2a, feasibility trial. Lancet Oncol 2023; 24:457-467. [PMID: 37062295 DOI: 10.1016/s1470-2045(23)00102-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Targeted real-time imaging during robot-assisted radical prostatectomy provides information on the localisation and extent of prostate cancer. We assessed the safety and feasibility of the prostate-specific membrane antigen (PSMA)-targeted fluorescent tracer OTL78 in patients with prostate cancer. METHODS In this single-arm, phase 2a, feasibility trial with an adaptive design was carried out in The Netherlands Cancer Institute, Netherlands. Male patients aged 18 years or older, with PSMA PET-avid prostate cancer with an International Society of Urological Pathology (ISUP) grade group of 2 or more, who were scheduled to undergo robot-assisted radical prostatectomy with or without extended pelvic lymph node dissection were eligible. All patients had a robot-assisted radical prostatectomy using OTL78. Based on timing and dose, patients received a single intravenous infusion of OTL78 (0·06 mg/kg 1-2 h before surgery [dose cohort 1], 0·03 mg/kg 1-2 h before surgery [dose cohort 2], or 0·03 mg/kg 24 h before surgery [dose cohort 3]). The primary outcomes, assessed in all enrolled patients, were safety and pharmacokinetics of OTL78. This study is completed and is registered in the European Trial Database, 2019-002393-31, and the International Clinical Trials Registry Platform, NL8552, and is completed. FINDINGS Between June 29, 2020, and April 1, 2021, 19 patients were screened for eligibility, 18 of whom were enrolled. The median age was 69 years (IQR 64-70) and median prostate-specific antigen concentration was 15 ng/mL (IQR 9·3-22·0). In 16 (89%) of 18 patients, robot-assisted radical prostatectomy was accompanied by an extended pelvic lymph node dissection. Three serious adverse events occurred in one (6%) patient: an infected lymphocele, a urosepsis, and an intraperitoneal haemorrhage. These adverse events were considered unrelated to the administration of OTL78 or intraoperative fluorescence imaging. No patient died, required a dose reduction, or required discontinuation due to drug-related toxicity. The dose-normalised maximum serum concentration (Cmax/dose) in patients was 84·1 ng/mL/mg for the 0·03 mg/kg dose and 79·6 ng/mL/mg for the 0·06 mg/kg dose, the half-life was 5·1 h for the 0·03 mg/kg dose and 4·7 h for the 0·06 mg/kg dose, the volume of distribution was 22·9 L for the 0·03 mg/kg dose and 19·5 L for the 0·06 mg/kg dose, and the clearance was 3·1 L/h for the 0·03 mg/kg dose and 3·0 L/h for the 0·06 mg/kg dose. INTERPRETATION This first-in-patient study showed that OTL78 was well tolerated and had the potential to improve prostate cancer detection. Optimal dosing was 0·03 mg/kg, 24 h preoperatively. PSMA-directed fluorescence imaging allowed real-time identification of visually occult prostate cancer and might help to achieve complete oncological resections. FUNDING On Target Laboratories.
Collapse
Affiliation(s)
- Judith A Stibbe
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Hilda A de Barros
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Daan G J Linders
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Shadhvi S Bhairosingh
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Elise M Bekers
- Department of Pathology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Pim J van Leeuwen
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | | | | | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands; Centre for Human Drug Research, Leiden, Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands; Department of Urology, Amsterdam University Medical Center, Amsterdam, Netherlands.
| |
Collapse
|
21
|
Verhoeven M, Handula M, van den Brink L, de Ridder CMA, Stuurman DC, Seimbille Y, Dalm SU. Pre- and Intraoperative Visualization of GRPR-Expressing Solid Tumors: Preclinical Profiling of Novel Dual-Modality Probes for Nuclear and Fluorescence Imaging. Cancers (Basel) 2023; 15:cancers15072161. [PMID: 37046825 PMCID: PMC10093582 DOI: 10.3390/cancers15072161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/21/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Image-guided surgery using a gastrin-releasing peptide receptor (GRPR)-targeting dual-modality probe could improve the accuracy of the resection of various solid tumors. The aim of this study was to further characterize our four previously developed GRPR-targeting dual-modality probes that vary in linker structures and were labeled with indium-111 and sulfo-cyanine 5. Cell uptake studies with GRPR-positive PC-3 cells and GRPR-negative NCI-H69 cells confirmed receptor specificity. Imaging and biodistribution studies at 4 and 24 h with 20 MBq/1 nmol [111In]In-12-15 were performed in nude mice bearing a PC-3 and NCI-H69 xenograft, and showed that the probe with only a pADA linker in the backbone had the highest tumor-to-organ ratios (T/O) at 24 h after injection (T/O > 5 for, e.g., prostate, muscle and blood). For this probe, a dose optimization study with three doses (0.75, 1.25 and 1.75 nmol; 20 MBq) revealed that the maximum image contrast was achieved with the lowest dose. Subsequently, the probe was successfully used for tumor excision in a simulated image-guided surgery setting. Moreover, it demonstrated binding to tissue sections of human prostate, breast and gastro-intestinal stromal tumors. In summary, our findings demonstrate that the developed dual-modality probe has the potential to aid in the complete surgical removal of GRPR-positive tumors.
Collapse
Affiliation(s)
- Marjolein Verhoeven
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Lilian van den Brink
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Corrina M. A. de Ridder
- Department of Experimental Urology, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Debra C. Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
| | - Simone U. Dalm
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
22
|
Meher N, Ashley GW, Bidkar AP, Dhrona S, Fong C, Fontaine SD, Beckford Vera DR, Wilson DM, Seo Y, Santi DV, VanBrocklin HF, Flavell RR. Prostate-Specific Membrane Antigen Targeted Deep Tumor Penetration of Polymer Nanocarriers. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50569-50582. [PMID: 36318757 PMCID: PMC9673064 DOI: 10.1021/acsami.2c15095] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/24/2022] [Indexed: 05/05/2023]
Abstract
Tumoral uptake of large-size nanoparticles is mediated by the enhanced permeability and retention (EPR) effect, with variable accumulation and heterogenous tumor tissue penetration depending on the tumor phenotype. The performance of nanocarriers via specific targeting has the potential to improve imaging contrast and therapeutic efficacy in vivo with increased deep tissue penetration. To address this hypothesis, we designed and synthesized prostate cancer-targeting starPEG nanocarriers (40 kDa, 15 nm), [89Zr]PEG-(DFB)3(ACUPA)1 and [89Zr]PEG-(DFB)1(ACUPA)3, with one or three prostate-specific membrane antigen (PSMA)-targeting ACUPA ligands. The in vitro PSMA binding affinity and in vivo pharmacokinetics of the targeted nanocarriers were compared with a nontargeted starPEG, [89Zr]PEG-(DFB)4, in PSMA+ PC3-Pip and PSMA- PC3-Flu cells, and xenografts. Increasing the number of ACUPA ligands improved the in vitro binding affinity of PEG-derived polymers to PC3-Pip cells. While both PSMA-targeted nanocarriers significantly improved tissue penetration in PC3-Pip tumors, the multivalent [89Zr]PEG-(DFB)1(ACUPA)3 showed a remarkably higher PC3-Pip/blood ratio and background clearance. In contrast, the nontargeted [89Zr]PEG-(DFB)4 showed low EPR-mediated accumulation with poor tumor tissue penetration. Overall, ACUPA conjugated targeted starPEGs significantly improve tumor retention with deep tumor tissue penetration in low EPR PC3-Pip xenografts. These data suggest that PSMA targeting with multivalent ACUPA ligands may be a generally applicable strategy to increase nanocarrier delivery to prostate cancer. These targeted multivalent nanocarriers with high tumor binding and low healthy tissue retention could be employed in imaging and therapeutic applications.
Collapse
Affiliation(s)
- Niranjan Meher
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - Gary W. Ashley
- ProLynx
Inc., San Francisco, California 94158, United States
| | - Anil P. Bidkar
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - Suchi Dhrona
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - Cyril Fong
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | | | - Denis R. Beckford Vera
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
| | - David M. Wilson
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Youngho Seo
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Daniel V. Santi
- ProLynx
Inc., San Francisco, California 94158, United States
| | - Henry F. VanBrocklin
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Robert R. Flavell
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, California 94143, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
- Department
of Pharmaceutical Chemistry, University
of California, San Francisco, California 94158-2517, United States
| |
Collapse
|
23
|
Yi X, Wang Z, Hu X, Yu A. Affinity probes based on small-molecule inhibitors for tumor imaging. Front Oncol 2022; 12:1028493. [PMID: 36387103 PMCID: PMC9647038 DOI: 10.3389/fonc.2022.1028493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/17/2022] [Indexed: 11/29/2022] Open
Abstract
Methods for molecular imaging of target areas, including optical imaging, radionuclide imaging, magnetic resonance imaging and other imaging technologies, are helpful for the early diagnosis and precise treatment of cancers. In addition to cancer management, small-molecule inhibitors are also used for developing cancer target probes since they act as the tight-binding ligands of overexpressed proteins in cancer cells. This review aims to summarize the structural designs of affinity probes based on small-molecule inhibitors from the aspects of the inhibitor, linker, dye and radionuclide, and discusses the influence of the modification of these structures on affinity and pharmacokinetics. We also present examples of inhibitor affinity probes in clinical applications, and these summaries will provide insights for future research and clinical translations.
Collapse
Affiliation(s)
| | | | - Xiang Hu
- *Correspondence: Aixi Yu, ; Xiang Hu,
| | - Aixi Yu
- *Correspondence: Aixi Yu, ; Xiang Hu,
| |
Collapse
|
24
|
Li Y, Duan X, Xu H, Zhang J, Zhou H, Zhang X, Zhang J, Yang Z, Hu Z, Zhang N, Tian J, Yang X. Optimization of ODAP-Urea-based dual-modality PSMA targeting probes for sequential PET-CT and optical imaging. Bioorg Med Chem 2022; 66:116810. [PMID: 35580538 DOI: 10.1016/j.bmc.2022.116810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 11/02/2022]
Abstract
Prostate-specific membrane antigen (PSMA) is emerging as a promising target to specifically image prostate cancer. Dual-modality probe combining radionuclide imaging and near-infrared fluorescence navigation targeting PSMA would enable both the preoperative staging and intraoperative detection of the tumor lesions. To overcome one of the key barriers for achieving high contrast imaging at both early and late time points, we optimized the pharmacokinetics of dual-modality probes based on oxalyldiaminopropionic acid-urea (ODAP-Urea) PSMA inhibitors recently developed. Four dual-modality probes with variable hydrophilicity were synthesized and evaluated. They displayed good optical properties (λem max = 835 nm, QY = 0.67%-1.50%), high affinity to PSMA (Ki = 2.09 ± 1.71-4.15 ± 2.20 nM) and PSMA specific cellular uptake (0.48 ± 0.01% - 0.64 ± 0.04% IA/105 LNCaP cells) upon labeled with 68Ga. In vivo studies showed that [68Ga]Ga-P3 exhibited an optimum pharmacokinetic property with high specific tumor uptake (SUVmax = 1.88 ± 0.36, at 1 h) in medium level PSMA expressing 22Rv1 tumor model and high tumor-to-muscle ratio (12.56 ± 2.63, at 1 h). Specific fluorescence imaging could also be achieved with high contrast for later time points (tumor-to-background ratio = 11.63 ± 4.16 at 24 h). This study demonstrates that ODAP-Urea-based P3 has the potential for PET imaging and intraoperative optical imaging of prostate cancer.
Collapse
Affiliation(s)
- Yuan Li
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China; NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing 100142, China
| | - Hongchuang Xu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Jingming Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Haoxi Zhou
- Department of Nuclear Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jinming Zhang
- Department of Nuclear Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zhi Yang
- NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Ning Zhang
- Translational Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing 100191, China.
| | - Xing Yang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China; NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing 100142, China.
| |
Collapse
|
25
|
Dual-Labelling Strategies for Nuclear and Fluorescence Molecular Imaging: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15040432. [PMID: 35455430 PMCID: PMC9028399 DOI: 10.3390/ph15040432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Molecular imaging offers the possibility to investigate biological and biochemical processes non-invasively and to obtain information on both anatomy and dysfunctions. Based on the data obtained, a fundamental understanding of various disease processes can be derived and treatment strategies can be planned. In this context, methods that combine several modalities in one probe are increasingly being used. Due to the comparably high sensitivity and provided complementary information, the combination of nuclear and optical probes has taken on a special significance. In this review article, dual-labelled systems for bimodal nuclear and optical imaging based on both modular ligands and nanomaterials are discussed. Particular attention is paid to radiometal-labelled molecules for single-photon emission computed tomography (SPECT) and positron emission tomography (PET) and metal complexes combined with fluorescent dyes for optical imaging. The clinical potential of such probes, especially for fluorescence-guided surgery, is assessed.
Collapse
|
26
|
Usama SM, Marker SC, Hernandez Vargas S, AghaAmiri S, Ghosh SC, Ikoma N, Tran Cao HS, Schnermann MJ, Azhdarinia A. Targeted Dual-Modal PET/SPECT-NIR Imaging: From Building Blocks and Construction Strategies to Applications. Cancers (Basel) 2022; 14:1619. [PMID: 35406390 PMCID: PMC8996983 DOI: 10.3390/cancers14071619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Molecular imaging is an emerging non-invasive method to qualitatively and quantitively visualize and characterize biological processes. Among the imaging modalities, PET/SPECT and near-infrared (NIR) imaging provide synergistic properties that result in deep tissue penetration and up to cell-level resolution. Dual-modal PET/SPECT-NIR agents are commonly combined with a targeting ligand (e.g., antibody or small molecule) to engage biomolecules overexpressed in cancer, thereby enabling selective multimodal visualization of primary and metastatic tumors. The use of such agents for (i) preoperative patient selection and surgical planning and (ii) intraoperative FGS could improve surgical workflow and patient outcomes. However, the development of targeted dual-modal agents is a chemical challenge and a topic of ongoing research. In this review, we define key design considerations of targeted dual-modal imaging from a topological perspective, list targeted dual-modal probes disclosed in the last decade, review recent progress in the field of NIR fluorescent probe development, and highlight future directions in this rapidly developing field.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Sierra C. Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Sukhen C. Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (N.I.); (H.S.T.C.)
| | - Hop S. Tran Cao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (N.I.); (H.S.T.C.)
| | - Martin J. Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| |
Collapse
|
27
|
Yuen R, West FG, Wuest F. Dual Probes for Positron Emission Tomography (PET) and Fluorescence Imaging (FI) of Cancer. Pharmaceutics 2022; 14:pharmaceutics14030645. [PMID: 35336019 PMCID: PMC8952779 DOI: 10.3390/pharmaceutics14030645] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 02/07/2023] Open
Abstract
Dual probes that possess positron emission tomography (PET) and fluorescence imaging (FI) capabilities are precision medicine tools that can be used to improve patient care and outcomes. Detecting tumor lesions using PET, an extremely sensitive technique, coupled with fluorescence-guided surgical resection of said tumor lesions can maximize the removal of cancerous tissue. The development of novel molecular probes is important for targeting different biomarkers as every individual case of cancer has different characteristics. This short review will discuss some aspects of dual PET/FI probes and explore the recently reported examples.
Collapse
Affiliation(s)
- Richard Yuen
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; (R.Y.); (F.G.W.)
| | - Frederick G. West
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; (R.Y.); (F.G.W.)
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Frank Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; (R.Y.); (F.G.W.)
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Oncology, University of Alberta—Cross Cancer Institute, Edmonton, AB T6G IZ2, Canada
- Correspondence:
| |
Collapse
|
28
|
Dell'Oglio P, van Willigen DM, van Oosterom MN, Bauwens K, Hensbergen F, Welling MM, van der Stadt H, Bekers E, Pool M, van Leeuwen P, Maurer T, van Leeuwen FWB, Buckle T. Feasibility of fluorescence imaging at microdosing using a hybrid PSMA tracer during robot-assisted radical prostatectomy in a large animal model. EJNMMI Res 2022; 12:14. [PMID: 35254544 PMCID: PMC8901828 DOI: 10.1186/s13550-022-00886-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/19/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND With the rise of prostate-specific membrane antigen (PSMA) radioguided surgery, which is performed using a microdosing regime, demand for visual target confirmation via fluorescence guidance is growing. While proven very effective for radiotracers, microdosing approaches the detection limit for fluorescence imaging. Thus, utility will be highly dependent on the tracer performance, the sensitivity of the fluorescence camera used, and the degree of background signal. Using a porcine model the ability to perform robot-assisted radical prostatectomy under fluorescence guidance using the bimodal or rather hybrid PSMA tracer (99mTc-EuK-(SO3)Cy5-mas3) was studied, while employing the tracer in a microdosing regime. This was followed by ex vivo evaluation in surgical specimens obtained from prostate cancer patients. RESULTS T50% blood and T50% urine were reached at 85 min and 390 min, in, respectively, blood and urine. Surgical fluorescence imaging allowed visualization of the prostate gland based on the basal PSMA-expression in porcine prostate. Together, in vivo visualization of the prostate and urinary excretion suggests at least an interval of > 7 h between tracer administration and surgery. Confocal microscopy of excised tissues confirmed tracer uptake in kidney and prostate, which was confirmed with PSMA IHC. No fluorescence was detected in other excised tissues. Tumor identification based on ex vivo fluorescence imaging of human prostate cancer specimens correlated with PSMA IHC. CONCLUSION Intraoperative PSMA-mediated fluorescence imaging with a microdosing approach was shown to be feasible. Furthermore, EuK-(SO3)Cy5-mas3 allowed tumor identification in human prostate samples, underlining the translational potential of this novel tracer. Trial registration Approval for use of biological material for research purposes was provided by the Translational Research Board of the Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital (NKI-AvL) under reference IRBm19-273 (22/10/2019).
Collapse
Affiliation(s)
- Paolo Dell'Oglio
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- Department of Urology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Danny M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - Fabian Hensbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Mick M Welling
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | | | - Elise Bekers
- Department of Pathology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Martin Pool
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pim van Leeuwen
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Tobias Maurer
- Martini-Klinik, Universit¨Atsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
- Department of Urology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Chen Y, Minn I, Rowe SP, Lisok A, Chatterjee S, Brummet M, Banerjee SR, Mease RC, Pomper MG. A Series of PSMA-Targeted Near-Infrared Fluorescent Imaging Agents. Biomolecules 2022; 12:biom12030405. [PMID: 35327597 PMCID: PMC8946146 DOI: 10.3390/biom12030405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
We have synthesized a series of 10 new, PSMA-targeted, near-infrared imaging agents intended for use in vivo for fluorescence-guided surgery (FGS). Compounds were synthesized from the commercially available amine-reactive active NHS ester of DyLight800. We altered the linker between the PSMA-targeting urea moiety and the fluorophore with a view to improve the pharmacokinetics. Chemical yields for the conjugates ranged from 51% to 86%. The Ki values ranged from 0.10 to 2.19 nM. Inclusion of an N-bromobenzyl substituent at the ε-amino group of lysine enhanced PSMA+ PIP tumor uptake, as did hydrophilic substituents within the linker. The presence of a polyethylene glycol chain within the linker markedly decreased renal uptake. In particular, DyLight800-10 demonstrated high specific uptake relative to background signal within kidney, confirmed by immunohistochemistry. These compounds may be useful for FGS in prostate, renal or other PSMA-expressing cancers.
Collapse
|
30
|
Eder AC, Matthias J, Schäfer M, Schmidt J, Steinacker N, Bauder-Wüst U, Domogalla LC, Roscher M, Haberkorn U, Eder M, Kopka K. A New Class of PSMA-617-Based Hybrid Molecules for Preoperative Imaging and Intraoperative Fluorescence Navigation of Prostate Cancer. Pharmaceuticals (Basel) 2022; 15:ph15030267. [PMID: 35337061 PMCID: PMC8954540 DOI: 10.3390/ph15030267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
The development of PSMA-targeting low-molecular-weight hybrid molecules aims at advancing preoperative imaging and accurate intraoperative fluorescence guidance for improved diagnosis and therapy of prostate cancer. In hybrid probe design, the major challenge is the introduction of a bulky dye to peptidomimetic core structures without affecting tumor-targeting properties and pharmacokinetic profiles. This study developed a novel class of PSMA-targeting hybrid molecules based on the clinically established theranostic agent PSMA-617. The fluorescent dye-bearing candidates of the strategically designed molecule library were evaluated in in vitro assays based on their PSMA-binding affinity and internalization properties to identify the most favorable hybrid molecule composition for the installation of a bulky dye. The library’s best candidate was realized with IRDye800CW providing the lead compound. Glu-urea-Lys-2-Nal-Chx-Lys(IRDye800CW)-DOTA (PSMA-927) was investigated in an in vivo proof-of-concept study, with compelling performance in organ distribution studies, PET/MRI and optical imaging, and with a strong PSMA-specific tumor uptake comparable to that of PSMA-617. This study provides valuable insights about the design of PSMA-targeting low-molecular-weight hybrid molecules, which enable further advances in the field of peptidomimetic hybrid molecule development.
Collapse
Affiliation(s)
- Ann-Christin Eder
- Department of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.S.); (J.S.); (U.B.-W.); (M.R.); (K.K.)
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (N.S.); (L.-C.D.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-761-270-74221
| | - Jessica Matthias
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany;
| | - Martin Schäfer
- Department of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.S.); (J.S.); (U.B.-W.); (M.R.); (K.K.)
| | - Jana Schmidt
- Department of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.S.); (J.S.); (U.B.-W.); (M.R.); (K.K.)
| | - Nils Steinacker
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (N.S.); (L.-C.D.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Ulrike Bauder-Wüst
- Department of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.S.); (J.S.); (U.B.-W.); (M.R.); (K.K.)
| | - Lisa-Charlotte Domogalla
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (N.S.); (L.-C.D.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Mareike Roscher
- Department of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.S.); (J.S.); (U.B.-W.); (M.R.); (K.K.)
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (N.S.); (L.-C.D.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Klaus Kopka
- Department of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (M.S.); (J.S.); (U.B.-W.); (M.R.); (K.K.)
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01069 Dresden, Germany
| |
Collapse
|
31
|
Identification of alternative protein targets of glutamate-ureido-lysine associated with PSMA tracer uptake in prostate cancer cells. Proc Natl Acad Sci U S A 2022; 119:2025710119. [PMID: 35064078 PMCID: PMC8795759 DOI: 10.1073/pnas.2025710119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2021] [Indexed: 12/21/2022] Open
Abstract
Glutamate-ureido-lysine (GUL) probes are specific for prostate-specific membrane antigen (PSMA), overexpressed by most prostate cancers. This antigen can be lost as the cancer progresses. Recent reports have indicated that GUL probes can still identify these PSMA-negative tumors, indicating that the expression of alternative PSMA-like proteins may change during disease progression. In this study we identified two such candidate protein targets, NAALADaseL and mGluR8, by using a combined computational chemistry, data mining, molecular biology, radiochemistry, and synthetic chemistry approach. This work consequently prepares the groundwork for developing specific probes that can identify this progression, indicates directions for neuroendocrine prostate cancer research, and highlights the utility of a multidisciplinary approach for the rapid identification of unidentified proteins interacting with diagnostic probes. Prostate-specific membrane antigen (PSMA) is highly overexpressed in most prostate cancers and is clinically visualized using PSMA-specific probes incorporating glutamate-ureido-lysine (GUL). PSMA is effectively absent from certain high-mortality, treatment-resistant subsets of prostate cancers, such as neuroendocrine prostate cancer (NEPC); however, GUL-based PSMA tracers are still reported to have the potential to identify NEPC metastatic tumors. These probes may bind unknown proteins associated with PSMA-suppressed cancers. We have identified the up-regulation of PSMA-like aminopeptidase NAALADaseL and the metabotropic glutamate receptors (mGluRs) in PSMA-suppressed prostate cancers and find that their expression levels inversely correlate with PSMA expression and are associated with GUL-based radiotracer uptake. Furthermore, we identify that NAALADaseL and mGluR expression correlates with a unique cell cycle signature. This provides an opportunity for the future study of the biology of NEPC and potential therapeutic directions. Computationally predicting that GUL-based probes bind well to these targets, we designed and synthesized a fluorescent PSMA tracer to investigate these proteins in vitro, where it shows excellent affinity for PSMA, NAALADaseL, and specific mGluRs associated with poor prognosis.
Collapse
|
32
|
Derks YHW, Rijpkema M, Amatdjais-Groenen HIV, Loeff CC, de Roode KE, Kip A, Laverman P, Lütje S, Heskamp S, Löwik DWPM. Strain-Promoted Azide-Alkyne Cycloaddition-Based PSMA-Targeting Ligands for Multimodal Intraoperative Tumor Detection of Prostate Cancer. Bioconjug Chem 2022; 33:194-205. [PMID: 34957825 PMCID: PMC8778659 DOI: 10.1021/acs.bioconjchem.1c00537] [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: 11/16/2021] [Revised: 12/15/2021] [Indexed: 01/18/2023]
Abstract
Strain-promoted azide-alkyne cycloaddition (SPAAC) is a straightforward and multipurpose conjugation strategy. The use of SPAAC to link different functional elements to prostate-specific membrane antigen (PSMA) ligands would facilitate the development of a modular platform for PSMA-targeted imaging and therapy of prostate cancer (PCa). As a first proof of concept for the SPAAC chemistry platform, we synthesized and characterized four dual-labeled PSMA ligands for intraoperative radiodetection and fluorescence imaging of PCa. Ligands were synthesized using solid-phase chemistry and contained a chelator for 111In or 99mTc labeling. The fluorophore IRDye800CW was conjugated using SPAAC chemistry or conventional N-hydroxysuccinimide (NHS)-ester coupling. Log D values were measured and PSMA specificity of these ligands was determined in LS174T-PSMA cells. Tumor targeting was evaluated in BALB/c nude mice with subcutaneous LS174T-PSMA and LS174T wild-type tumors using μSPECT/CT imaging, fluorescence imaging, and biodistribution studies. SPAAC chemistry increased the lipophilicity of the ligands (log D range: -2.4 to -4.4). In vivo, SPAAC chemistry ligands showed high and specific accumulation in s.c. LS174T-PSMA tumors up to 24 h after injection, enabling clear visualization using μSPECT/CT and fluorescence imaging. Overall, no significant differences between the SPAAC chemistry ligands and their NHS-based counterparts were found (2 h p.i., p > 0.05), while 111In-labeled ligands outperformed the 99mTc ligands. Here, we demonstrate that our newly developed SPAAC-based PSMA ligands show high PSMA-specific tumor targeting. The use of click chemistry in PSMA ligand development opens up the opportunity for fast, efficient, and versatile conjugations of multiple imaging moieties and/or drugs.
Collapse
Affiliation(s)
- Yvonne H. W. Derks
- Department
of Medical Imaging, Nuclear Medicine, Radboud
university medical center, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Mark Rijpkema
- Department
of Medical Imaging, Nuclear Medicine, Radboud
university medical center, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | | | - Cato C. Loeff
- Department
of Medical Imaging, Nuclear Medicine, Radboud
university medical center, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Kim E. de Roode
- Organic
Chemistry, Radboud University Nijmegen,
Institute for Molecules and Materials, 6525XZ Nijmegen, The Netherlands
| | - Annemarie Kip
- Department
of Medical Imaging, Nuclear Medicine, Radboud
university medical center, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Peter Laverman
- Department
of Medical Imaging, Nuclear Medicine, Radboud
university medical center, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Susanne Lütje
- Department
of Nuclear Medicine, University Hospital
Bonn, 53127 Bonn, Germany
| | - Sandra Heskamp
- Department
of Medical Imaging, Nuclear Medicine, Radboud
university medical center, Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Dennis W. P. M. Löwik
- Organic
Chemistry, Radboud University Nijmegen,
Institute for Molecules and Materials, 6525XZ Nijmegen, The Netherlands
| |
Collapse
|
33
|
Ariztia J, Solmont K, Moïse NP, Specklin S, Heck MP, Lamandé-Langle S, Kuhnast B. PET/Fluorescence Imaging: An Overview of the Chemical Strategies to Build Dual Imaging Tools. Bioconjug Chem 2022; 33:24-52. [PMID: 34994545 DOI: 10.1021/acs.bioconjchem.1c00503] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular imaging is a biomedical research discipline that has quickly emerged to afford the observation, characterization, monitoring, and quantification of biomarkers and biological processes in living organism. It covers a large array of imaging techniques, each of which provides anatomical, functional, or metabolic information. Multimodality, as the combination of two or more of these techniques, has proven to be one of the best options to boost their individual properties, hence offering unprecedented tools for human health. In this review, we will focus on the combination of positron emission tomography and fluorescence imaging from the specific perspective of the chemical synthesis of dual imaging agents. Based on a detailed analysis of the literature, this review aims at giving a comprehensive overview of the chemical strategies implemented to build adequate imaging tools considering radiohalogens and radiometals as positron emitters, fluorescent dyes mostly emitting in the NIR window and all types of targeting vectors.
Collapse
Affiliation(s)
- Julen Ariztia
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Kathleen Solmont
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | | | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Marie Pierre Heck
- Université Paris-Saclay, INRAE, Département Médicaments et Technologies pour la santé (DMTS), SCBM, 91191, Gif-sur-Yvette cedex, France
| | | | - Bertrand Kuhnast
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| |
Collapse
|
34
|
Declerck NB, Mateusiak L, Hernot S. Design and Validation of Site-Specifically Labeled Single-Domain Antibody-Based Tracers for in Vivo Fluorescence Imaging and Image-Guided Surgery. Methods Mol Biol 2022; 2446:395-407. [PMID: 35157285 DOI: 10.1007/978-1-0716-2075-5_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Near-infrared fluorescence molecular imaging has become an established preclinical technique to investigate molecular processes in vivo and to study novel therapies. Furthermore, fluorescence molecular imaging is gaining significant interest from clinicians as an intra-operative guidance tool. This technique makes use of targeted fluorescent tracers as contrast agents that recognize specific biomarkers expressed at the site of disease. Single-domain antibodies have shown to possess excellent properties for in vivo imaging in comparison to conventional antibodies. In this chapter, we describe a method for site-specific conjugation of a near-infrared fluorophore to single-domain antibodies by exploiting cysteine-maleimide chemistry. As opposed to random conjugation, site-specific conjugation results in a homogenously labeled fluorescent tracer and avoids inference with antigen binding.
Collapse
Affiliation(s)
- Noemi B Declerck
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lukasz Mateusiak
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophie Hernot
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
35
|
Buckle T, van Willigen DM, Welling MM, van Leeuwen FW. Pre-clinical development of fluorescent tracers and translation towards clinical application. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
36
|
Neels OC, Kopka K, Liolios C, Afshar-Oromieh A. Radiolabeled PSMA Inhibitors. Cancers (Basel) 2021; 13:6255. [PMID: 34944875 PMCID: PMC8699044 DOI: 10.3390/cancers13246255] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
PSMA has shown to be a promising target for diagnosis and therapy (theranostics) of prostate cancer. We have reviewed developments in the field of radio- and fluorescence-guided surgery and targeted photodynamic therapy as well as multitargeting PSMA inhibitors also addressing albumin, GRPr and integrin αvβ3. An overview of the regulatory status of PSMA-targeting radiopharmaceuticals in the USA and Europe is also provided. Technical and quality aspects of PSMA-targeting radiopharmaceuticals are described and new emerging radiolabeling strategies are discussed. Furthermore, insights are given into the production, application and potential of alternatives beyond the commonly used radionuclides for radiolabeling PSMA inhibitors. An additional refinement of radiopharmaceuticals is required in order to further improve dose-limiting factors, such as nephrotoxicity and salivary gland uptake during endoradiotherapy. The improvement of patient treatment achieved by the advantageous combination of radionuclide therapy with alternative therapies is also a special focus of this review.
Collapse
Affiliation(s)
- Oliver C. Neels
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328 Dresden, Germany;
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328 Dresden, Germany;
- Faculty of Chemistry and Food Chemistry, School of Science, Technical University Dresden, Mommsenstrasse 4, 01062 Dresden, Germany
| | - Christos Liolios
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National & Kapodistrian University of Athens, Zografou, 15771 Athens, Greece;
- INRASTES, Radiochemistry Laboratory, NCSR “Demokritos”, Ag. Paraskevi Attikis, 15310 Athens, Greece
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Bern University Hospital (Inselspital), Freiburgstrasse 18, 3010 Bern, Switzerland;
| |
Collapse
|
37
|
Petrov SA, Zyk NY, Machulkin AE, Beloglazkina EK, Majouga AG. PSMA-targeted low-molecular double conjugates for diagnostics and therapy. Eur J Med Chem 2021; 225:113752. [PMID: 34464875 DOI: 10.1016/j.ejmech.2021.113752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022]
Abstract
This review presents data on dual conjugates of therapeutic and diagnostic action for targeted delivery to prostate cancer cells. The works of the last ten years on this topic were analyzed. The mail attention focuses on low-molecular-weight conjugates directed to the prostate-specific membrane antigen (PSMA); the comparison of high and low molecular weight PSMA-targeted conjugates was made. The considered conjugates were divided in the review into two main classes: diagnostic bimodal conjugates (which are containing two fragments for different types of diagnostics), theranostic conjugates (containing both therapeutic and diagnostic agents); also bimodal high molecular weight therapeutic conjugates containing two therapeutic agents are briefly discussed. The data of in vitro and in vivo studies for PSMA-targeted double conjugates available by the beginning of 2021 have been analyzed.
Collapse
Affiliation(s)
- Stanislav A Petrov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Nikolay Y Zyk
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | | | | | - Alexander G Majouga
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia; Laboratory of Biomedical Nanomaterials, National University of Science and Technology MISiS, Moscow, Russia; Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| |
Collapse
|
38
|
Eder AC, Schäfer M, Schmidt J, Bauder-Wüst U, Roscher M, Leotta K, Haberkorn U, Kopka K, Eder M. Rational Linker Design to Accelerate Excretion and Reduce Background Uptake of Peptidomimetic PSMA-Targeting Hybrid Molecules. J Nucl Med 2021; 62:1461-1467. [PMID: 33741642 PMCID: PMC8724895 DOI: 10.2967/jnumed.120.248443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 01/13/2021] [Indexed: 11/16/2022] Open
Abstract
The evolution of peptidomimetic hybrid molecules for preoperative imaging and guided surgery targeting the prostate-specific membrane antigen (PSMA) significantly progressed over the past few years, and some approaches are currently being evaluated for further clinical translation. However, accumulation in nonmalignant tissue such as kidney, bladder, spleen, or liver might limit tumor-to-background contrast for precise lesion delineation, particularly in a surgical setting. To overcome these limitations, a rational linker design aims at the development of a second generation of PSMA-11-based hybrid molecules with an enhanced pharmacokinetic profile and improved imaging contrast. Methods: A selection of rationally designed linkers was introduced to the PSMA-targeting hybrid molecule Glu-urea-Lys-HBED-CC-IRDye800CW, resulting in a second-generation peptidomimetic hybrid molecule library. The biologic properties were investigated in cell-based assays. In a preclinical proof-of-concept study with the radionuclide 68Ga, the impact of the modifications was evaluated by determination of specific tumor uptake, pharmacokinetics, and fluorescence imaging in tumor-bearing mice. Results: The modified hybrid molecules carrying various selected linkers revealed high PSMA-specific binding affinity and effective internalization. The highest tumor-to-background contrast of all modifications investigated was identified for the introduction of a histidine- (H) and glutamic acid (E)-containing linker ((HE)3-linker) between the PSMA-binding motif and the chelator. In comparison to the parental core structure, uptake in nonmalignant tissue was significantly reduced to a minimum, as exemplified by an 11-fold reduced spleen uptake from 38.12 ± 14.62 percentage injected dose (%ID)/g to 3.47 ± 1.39 %ID/g (1 h after injection). The specific tumor uptake of this compound (7.59 ± 0.95 %ID/g, 1 h after injection) was detected to be significantly higher than that of the parental tracer PSMA-11. These findings confirmed by PET and fluorescence imaging are accompanied by an enhanced pharmacokinetic profile with accelerated background clearance at early time points after injection. Conclusion: The novel generation of PSMA-targeting hybrid molecules reveals fast elimination, reduced background organ enrichment, and high PSMA-specific tumor uptake meeting the key demands for potent tracers in nuclear medicine and fluorescence-guided surgery. The approach's efficacy in improving the pharmacokinetic profile highlights the strengths of rational linker design as a powerful tool in strategic hybrid-molecule development.
Collapse
Affiliation(s)
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jana Schmidt
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Bauder-Wüst
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mareike Roscher
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Leotta
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DFKZ), Heidelberg, Germany; and
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DFKZ), Heidelberg, Germany; and
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DFKZ), Heidelberg, Germany; and
| |
Collapse
|
39
|
Synthesis, Characterization and In Vitro Evaluation of Hybrid Monomeric Peptides Suited for Multimodal Imaging by PET/OI: Extending the Concept of Charge-Cell Binding Correlation. Pharmaceuticals (Basel) 2021; 14:ph14100989. [PMID: 34681213 PMCID: PMC8541144 DOI: 10.3390/ph14100989] [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: 09/06/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
In the context of hybrid multimodal imaging agents for gastrin releasing peptide receptor (GRPR) targeting, a correlation between the net charge and the receptor affinity of the agents was recently found. In particular, a decrease in in vitro GRPR binding affinity was observed in case of an increasing number of negative charges for dually labeled GRPR-specific peptide dimers suited for positron emission tomography and optical imaging (PET/OI). This adverse influence of anionic charges could be in part compensated by a higher valency of peptide multimerization. However, it remains unknown whether this adverse effect of anionic charges is limited to peptide multimers or if it is also found or even more pronounced when GRPR-specific peptide monomers are dually labeled with fluorescent dye and chelating agent/radionuclide. Moreover, it would be important to know if this effect is limited to GRPR-specific agents only or if these observations also apply to other dually labeled peptides binding to other receptor types. To address these questions, we synthesized hybrid labels, comprising a chelator, different fluorescent dyes carrying different net charges and a functional group for bioconjugation and introduced them into different peptides, specifically targeting the GRPR, the melanocortin-1 receptor (MC1R) and integrin αvβ3. The synthesized conjugates were evaluated with regard to their chemical, radiochemical, photophysical and receptor affinity properties. It was found that neither the 68Ga-radiolabeling nor the fluorescence characteristics of the dyes were altered by the conjugation of the MIUs to the peptides. Further, it was confirmed that the net number of anionic charges has a negative effect on the GRPR-binding affinity of the GRPR-targeting MIU-peptide monomer conjugates and that this same effect was also found to the same extent for the other receptor systems studied.
Collapse
|
40
|
Chen H, Cai P, Feng Y, Sun Z, Wang Y, Chen Y, Zhang W, Liu N, Zhou Z. In vitro and in vivo comparative study of a novel 68Ga-labeled PSMA-targeted inhibitor and 68Ga-PSMA-11. Sci Rep 2021; 11:19122. [PMID: 34580375 PMCID: PMC8476564 DOI: 10.1038/s41598-021-98555-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
68Ga-radiolabeled small molecules that specifically target prostate-specific membrane antigen (PSMA) have been extensively investigated, and some of these tracers have been used in the diagnosis of prostate cancer via 68Ga-positron emission tomography (68Ga-PET). Nevertheless, current 68Ga-labeled radiotracers show only fair detection rates for metastatic prostate cancer lesions, especially those with lower levels of prostate specific antigen (PSA), which often occurs in the biochemical recurrence of prostate cancer. The goal of this study was to design and synthesize a new PSMA-targeted radiotracer, 68Ga-SC691, with high affinity for prostate cancer cells and excellent pharmacokinetics. To this end, structural optimization was carried out on the bifunctional group, target motif, and linker while the high affinity targeting scaffold remained. To explore its potential in the clinic, a comparative study was further performed in vitro and in vivo between 68Ga-SC691 and 68Ga-PSMA-11, a clinically approved tracer for PSMA-positive prostate cancer. SC691 was radiolabeled to provide 68Ga-SC691 in 99% radiolabeling yield under mild conditions. High uptake and a high internalization ratio into LNCaP cells were observed in in vitro studies. In vivo studies showed that 68Ga-SC691 had favorable biodistribution properties and could specifically accumulate on PSMA-positive LNCaP xenografts visualized by micro-PET/CT. This radiotracer showed excellent PET imaging quality and comparable, if not higher, uptake in LNCaP xenografts than 68Ga-PSMA-11.
Collapse
Affiliation(s)
- Huanyu Chen
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
| | - Ping Cai
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Yue Feng
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
| | - Zhanliang Sun
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
| | - Yinwen Wang
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Yue Chen
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
| | - Wei Zhang
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
| | - Nan Liu
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China. .,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China. .,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.
| | - Zhijun Zhou
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China. .,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China. .,Academician (Expert) Workstation of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China. .,Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
| |
Collapse
|
41
|
Stemler T, Hoffmann C, Hierlmeier IM, Maus S, Krause E, Ezziddin S, Jung G, Bartholomä MD. A Structure-Activity Relationship Study of Bimodal BODIPY-Labeled PSMA-Targeting Bioconjugates. ChemMedChem 2021; 16:2535-2545. [PMID: 33905162 PMCID: PMC8453963 DOI: 10.1002/cmdc.202100210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Indexed: 12/15/2022]
Abstract
The aim of this study was to identify a high-affinity BODIPY peptidomimetic that targets the prostate-specific membrane antigen (PSMA) as a potential bimodal imaging probe for prostate cancer. For the structure-activity study, several BODIPY (difluoroboron dipyrromethene) derivatives with varying spacers between the BODIPY dye and the PSMA Glu-CO-Lys binding motif were prepared. Corresponding affinities were determined by competitive binding assays in PSMA-positive LNCaP cells. One compound was identified with comparable affinity (IC50 =21.5±0.1 nM) to Glu-CO-Lys-Ahx-HBED-CC (PSMA-11) (IC50 =18.4±0.2 nM). Radiolabeling was achieved by Lewis-acid-mediated 19 F/18 F exchange in moderate molar activities (∼0.7 MBq nmol-1 ) and high radiochemical purities (>99 %) with mean radiochemical yields of 20-30 %. Cell internalization of the 18 F-labeled high-affinity conjugate was demonstrated in LNCaP cells showing gradual increasing PSMA-mediated internalization over time. By fluorescence microscopy, localization of the high-affinity BODIPY-PSMA conjugate was found in the cell membrane at early time points and also in subcellular compartments at later time points. In summary, a high-affinity BODIPY-PSMA conjugate has been identified as a suitable candidate for the development of PSMA-specific dual-imaging agents.
Collapse
Affiliation(s)
- Tobias Stemler
- Department of Nuclear MedicineSaarland University – Medical CenterKirrbergerstrasse66421HomburgGermany
| | - Caroline Hoffmann
- Department of Biophysical ChemistrySaarland UniversityCampus B2 266123SaarbrückenGermany
| | - Ina M. Hierlmeier
- Department of Nuclear MedicineSaarland University – Medical CenterKirrbergerstrasse66421HomburgGermany
| | - Stephan Maus
- Department of Nuclear MedicineSaarland University – Medical CenterKirrbergerstrasse66421HomburgGermany
| | - Elmar Krause
- Department of Cellular NeurophysiologyCenter for Integrative Physiology and Molecular Medicine (CIPMM)Saarland UniversityKirrbergerstrasse66421HomburgGermany
| | - Samer Ezziddin
- Department of Nuclear MedicineSaarland University – Medical CenterKirrbergerstrasse66421HomburgGermany
| | - Gregor Jung
- Department of Biophysical ChemistrySaarland UniversityCampus B2 266123SaarbrückenGermany
| | - Mark D. Bartholomä
- Department of Nuclear MedicineSaarland University – Medical CenterKirrbergerstrasse66421HomburgGermany
| |
Collapse
|
42
|
Cordonnier A, Boyer D, Besse S, Valleix R, Mahiou R, Quintana M, Briat A, Benbakkar M, Penault-Llorca F, Maisonial-Besset A, Maunit B, Tarrit S, Vivier M, Witkowski T, Mazuel L, Degoul F, Miot-Noirault E, Chezal JM. Synthesis and in vitro preliminary evaluation of prostate-specific membrane antigen targeted upconversion nanoparticles as a first step towards radio/fluorescence-guided surgery of prostate cancer. J Mater Chem B 2021; 9:7423-7434. [PMID: 34373887 DOI: 10.1039/d1tb00777g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the last decade, upconversion nanoparticles (UCNP) have been widely investigated in nanomedicine due to their high potential as imaging agents in the near-infrared (NIR) optical window of biological tissues. Here, we successfully develop active targeted UCNP as potential probes for dual NIR-NIR fluorescence and radioactive-guided surgery of prostate-specific membrane antigen (PSMA)(+) prostate cancers. We designed a one-pot thermolysis synthesis method to obtain oleic acid-coated spherical NaYF4:Yb,Tm@NaYF4 core/shell UCNP with narrow particle size distribution (30.0 ± 0.1 nm, as estimated by SAXS analysis) and efficient upconversion luminescence. Polyethylene glycol (PEG) ligands bearing different anchoring groups (phosphate, bis- and tetra-phosphonate-based) were synthesized and used to hydrophilize the UCNP. DLS studies led to the selection of a tetra-phosphonate PEG(2000) ligand affording water-dispersible UCNP with sustained colloidal stability in several aqueous media. PSMA-targeting ligands (i.e., glutamate-urea-lysine derivatives called KuEs) and fluorescent or radiolabelled prosthetic groups were grafted onto the UCNP surface by strain-promoted azide-alkyne cycloaddition (SPAAC). These UCNP, coated with 10 or 100% surface density of KuE ligands, did not induce cytotoxicity over 24 h incubation in LNCaP-Luc or PC3-Luc prostate cancer cell lines or in human fibroblasts for any of the concentrations evaluated. Competitive binding assays and flow cytometry demonstrated the excellent affinity of UCNP@KuE for PSMA-positive LNCaP-Luc cells compared with non-targeted UCNP@CO2H. Furthermore, the binding of UCNP@KuE to prostate tumour cells was positively correlated with the surface density of PSMA-targeting ligands and maintained after 125I-radiolabelling. Finally, a preliminary biodistribution study in LNCaP-Luc-bearing mice demonstrated the radiochemical stability of non-targeted [125I]UCNP paving the way for future in vivo assessments.
Collapse
Affiliation(s)
- Axel Cordonnier
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France. and Université Clermont Auvergne, Clermont Auvergne INP, CNRS, ICCF, F-63000 Clermont-Ferrand, France
| | - Damien Boyer
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, ICCF, F-63000 Clermont-Ferrand, France
| | - Sophie Besse
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Rodolphe Valleix
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, ICCF, F-63000 Clermont-Ferrand, France
| | - Rachid Mahiou
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, ICCF, F-63000 Clermont-Ferrand, France
| | - Mercedes Quintana
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Arnaud Briat
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Mhammed Benbakkar
- Université Clermont Auvergne, CNRS, Laboratoire Magmas et Volcans, UMR 6524, F-63000 Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France. and Department of Pathology and Biopathology, Jean Perrin Comprehensive Cancer Centre, Clermont-Ferrand, France
| | - Aurélie Maisonial-Besset
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Benoit Maunit
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Sébastien Tarrit
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Magali Vivier
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Tiffany Witkowski
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Leslie Mazuel
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Françoise Degoul
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Elisabeth Miot-Noirault
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| | - Jean-Michel Chezal
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
43
|
Hernandez Vargas S, Lin C, Tran Cao HS, Ikoma N, AghaAmiri S, Ghosh SC, Uselmann AJ, Azhdarinia A. Receptor-Targeted Fluorescence-Guided Surgery With Low Molecular Weight Agents. Front Oncol 2021; 11:674083. [PMID: 34277418 PMCID: PMC8279813 DOI: 10.3389/fonc.2021.674083] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer surgery remains the primary treatment option for most solid tumors and can be curative if all malignant cells are removed. Surgeons have historically relied on visual and tactile cues to maximize tumor resection, but clinical data suggest that relapse occurs partially due to incomplete cancer removal. As a result, the introduction of technologies that enhance the ability to visualize tumors in the operating room represents a pressing need. Such technologies have the potential to revolutionize the surgical standard-of-care by enabling real-time detection of surgical margins, subclinical residual disease, lymph node metastases and synchronous/metachronous tumors. Fluorescence-guided surgery (FGS) in the near-infrared (NIRF) spectrum has shown tremendous promise as an intraoperative imaging modality. An increasing number of clinical studies have demonstrated that tumor-selective FGS agents can improve the predictive value of fluorescence over non-targeted dyes. Whereas NIRF-labeled macromolecules (i.e., antibodies) spearheaded the widespread clinical translation of tumor-selective FGS drugs, peptides and small-molecules are emerging as valuable alternatives. Here, we first review the state-of-the-art of promising low molecular weight agents that are in clinical development for FGS; we then discuss the significance, application and constraints of emerging tumor-selective FGS technologies.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Therapeutics & Pharmacology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | | | - Hop S Tran Cao
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naruhiko Ikoma
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sukhen C Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Therapeutics & Pharmacology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
44
|
Martin KE, Cosby AG, Boros E. Multiplex and In Vivo Optical Imaging of Discrete Luminescent Lanthanide Complexes Enabled by In Situ Cherenkov Radiation Mediated Energy Transfer. J Am Chem Soc 2021; 143:9206-9214. [PMID: 34114809 DOI: 10.1021/jacs.1c04264] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recently, we pioneered the application of Cherenkov radiation (CR) of radionuclides for the in situ excitation of discrete Eu(III) and Tb(III) complexes. CR is produced by isotopes decaying under emission of charged particles in dielectric media and exhibits a maximum intensity below 400 nm. We have demonstrated that luminescent lanthanide antenna complexes are ideal acceptors for Cherenkov radiation-mediated energy transfer (CRET). Here, we develop and assess peptide-functionalized Tb(III) and Eu(III) complexes in conjunction with CRET excitation by the positron emissive radioisotope 18F for simultaneous, multiplexed imaging and in vivo optical imaging. This work demonstrates, for the first time, that the detection of the luminescence emission of a discrete Eu(III) complex in vivo is feasible. Our results open possibilities for discrete luminescent lanthanide complexes to be used as diagnostic, optical tools for the intrasurgical guidance of tumor resection.
Collapse
Affiliation(s)
- Kirsten E Martin
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Alexia G Cosby
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| |
Collapse
|
45
|
Collamati F, van Oosterom MN, Hadaschik BA, Fragoso Costa P, Darr C. Beta radioguided surgery: towards routine implementation? THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2021; 65:229-243. [PMID: 34014062 DOI: 10.23736/s1824-4785.21.03358-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION In locally or locally advanced solid tumors, surgery still remains a fundamental treatment method. However, conservative resection is associated with high collateral damage and functional limitations of the patient. Furthermore, the presence of residual tumor tissue following conservative surgical treatment is currently a common cause of locally recurrent cancer or of distant metastases. Reliable intraoperative detection of small cancerous tissue would allow surgeons to selectively resect malignant areas: this task can be achieved by means of image-guided surgery, such as beta radioguided surgery (RGS). EVIDENCE ACQUISITION In this paper, a comprehensive review of beta RGS is given, starting from the physical principles that differentiate beta from gamma radiation, that has already its place in nuclear medicine current practice. Also, the recent clinical feasibility of using Cerenkov radiation is discussed. EVIDENCE SYNTHESIS Despite being first proposed several decades ago, only in the last years a remarkable interest in beta RGS has been observed, probably driven by the diffusion of PET radio tracers. Today several different approaches are being pursued to assess the effectiveness of such a technique, including both beta+ and beta- emitting radiopharmaceuticals. CONCLUSIONS Beta RGS shows some peculiarities that can present it as a very promising complementary technique to standard procedures. Good results are being obtained in several tests, both ex vivo and in vivo. This might however be the time to initiate the trials to demonstrate the real clinical value of these technologies with seemingly clear potential.
Collapse
Affiliation(s)
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Boris A Hadaschik
- Department of Urology, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Pedro Fragoso Costa
- German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany.,Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Christopher Darr
- Department of Urology, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| |
Collapse
|
46
|
Matthias J, Engelhardt J, Schäfer M, Bauder-Wüst U, Meyer PT, Haberkorn U, Eder M, Kopka K, Hell SW, Eder AC. Cytoplasmic Localization of Prostate-Specific Membrane Antigen Inhibitors May Confer Advantages for Targeted Cancer Therapies. Cancer Res 2021; 81:2234-2245. [PMID: 33622696 DOI: 10.1158/0008-5472.can-20-1624] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 12/29/2020] [Accepted: 02/04/2021] [Indexed: 11/16/2022]
Abstract
Targeted imaging and therapy approaches based on novel prostate-specific membrane antigen (PSMA) inhibitors have fundamentally changed the treatment regimen of prostate cancer. However, the exact mechanism of PSMA inhibitor internalization has not yet been studied, and the inhibitors' subcellular fate remains elusive. Here, we investigated the intracellular distribution of peptidomimetic PSMA inhibitors and of PSMA itself by stimulated emission depletion (STED) nanoscopy, applying a novel nonstandard live cell staining protocol. Imaging analysis confirmed PSMA cluster formation at the cell surface of prostate cancer cells and clathrin-dependent endocytosis of PSMA inhibitors. Following the endosomal pathway, PSMA inhibitors accumulated in prostate cancer cells at clinically relevant time points. In contrast with PSMA itself, PSMA inhibitors were found to eventually distribute homogeneously in the cytoplasm, a molecular condition that promises benefits for treatment as cytoplasmic and in particular perinuclear enrichment of the radionuclide carriers may better facilitate the radiation-mediated damage of cancerous cells. This study is the first to reveal the subcellular fate of PSMA/PSMA inhibitor complexes at the nanoscale and aims to inspire the development of new approaches in the field of prostate cancer research, diagnostics, and therapeutics. SIGNIFICANCE: This study uses STED fluorescence microscopy to reveal the subcellular fate of PSMA/PSMA inhibitor complexes near the molecular level, providing insights of great clinical interest and suggestive of advantageous targeted therapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2234/F1.large.jpg.
Collapse
Affiliation(s)
- Jessica Matthias
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg, Germany.,Helmholtz International Graduate School, German Cancer Research Center, Heidelberg, Germany
| | - Johann Engelhardt
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center, Heidelberg, Germany
| | - Ulrike Bauder-Wüst
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center, Heidelberg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Radiopharmaceutical Development, German Cancer Consortium, Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Radiopharmaceutical Development, German Cancer Consortium, Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, Heidelberg, Germany
| | - Stefan W Hell
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg, Germany.,Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ann-Christin Eder
- Helmholtz International Graduate School, German Cancer Research Center, Heidelberg, Germany. .,Division of Radiopharmaceutical Chemistry, German Cancer Research Center, Heidelberg, Germany.,Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Radiopharmaceutical Development, German Cancer Consortium, Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
47
|
18F-fluorodeoxyglucose positron emission tomography-computed tomography for assessing organ distribution of stressed red blood cells in mice. Sci Rep 2021; 11:2505. [PMID: 33510312 PMCID: PMC7844045 DOI: 10.1038/s41598-021-82100-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/13/2021] [Indexed: 02/04/2023] Open
Abstract
Red blood cells (RBCs) stressed by high temperature are similar to senescent or damaged RBCs in pathological conditions. RBCs can be efficiently labelled with 18F-fluorodeoxyglucose (FDG). The aim of this study was to assess stressed RBCs erythrophagocytosis and organ distribution in vivo with the application of 18F-FDG PET/CT. RBCs were induced under high temperature (48 °C) to prepare stressed RBCs. Fluorescence-activated cell sorting (FACS) was used to analyse reactive oxygen species (ROS) generation, intracellular Ca2+ concentration and membrane phosphatidylserine (PS) externalization of RBCs. 18F-FDG was used to label RBCs and assess the erythrophagocytosis. Finally, 18F-FDG PET/CT was applied to reveal and measure the organ distribution of stressed RBCs in mice. Compared with untreated RBCs, stressed RBCs decreased in cell volume and increased in ROS level, intracellular Ca2+ concentration, and PS exposure. RBCs could be labelled by 18F-FDG. Stressed RBCs tended to be phagocytosed by macrophages via assessment of FACS and radioactivity. 18F-FDG PET/CT imaging showed that stressed RBCs were mainly trapped in spleen, while untreated RBCs remained in circulation system. Thus, stressed RBCs can be effectively labelled by 18F-FDG and tend to be trapped in spleen of mice as assessed by PET/CT.
Collapse
|
48
|
Rietbergen DD, VAN Oosterom MN, Kleinjan GH, Brouwer OR, Valdes-Olmos RA, VAN Leeuwen FW, Buckle T. Interventional nuclear medicine: a focus on radioguided intervention and surgery. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2021; 65:4-19. [PMID: 33494584 DOI: 10.23736/s1824-4785.21.03286-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Within interventional nuclear medicine (iNM) a prominent role is allocated for the sub-discipline of radioguided surgery. Unique for this discipline is the fact that an increasing number of clinical indications (e.g. lymphatic mapping, local tumor demarcation and/or tumor receptor targeted applications) have been adopted into routine care. The clinical integration is further strengthened by technical innovations in chemistry and engineering that enhance the translational potential of radioguided procedures in iNM. Together, these features not only ensure ongoing expansion of iNM but also warrant a lasting clinical impact for the sub-discipline of radioguided surgery.
Collapse
Affiliation(s)
- Daphne D Rietbergen
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthias N VAN Oosterom
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, the Netherlands.,Department of Urology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Gijs H Kleinjan
- Department of Urology, Leiden University Medical Center, Leiden, the Netherlands
| | - Oscar R Brouwer
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, the Netherlands.,Department of Urology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Renato A Valdes-Olmos
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Fijs W VAN Leeuwen
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, the Netherlands.,Department of Urology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Tessa Buckle
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, the Netherlands - .,Department of Urology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| |
Collapse
|
49
|
The PSMA-11-derived hybrid molecule PSMA-914 specifically identifies prostate cancer by preoperative PET/CT and intraoperative fluorescence imaging. Eur J Nucl Med Mol Imaging 2021; 48:2057-2058. [PMID: 33486568 PMCID: PMC8113185 DOI: 10.1007/s00259-020-05184-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/27/2020] [Indexed: 10/28/2022]
|
50
|
PET Radiochemistry. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|