1
|
Tsuchihashi S, Nakashima K, Watanabe H, Ono M. Development of Novel Gastrin-Releasing Peptide Receptor-Targeted Radioligand with Albumin Binder to Improve Accumulation in Tumor. ACS Med Chem Lett 2025; 16:797-803. [PMID: 40365411 PMCID: PMC12067139 DOI: 10.1021/acsmedchemlett.5c00032] [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: 01/15/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Gastrin-releasing peptide receptor (GRPR) is a promising target for cancer radiotheranostics combining nuclear imaging with targeted radionuclide therapy. Improving the accumulation of radioligands in tumors by introducing an albumin binder (ALB) is useful to promote the efficacy of radiotheranostics. In this study, we designed and synthesized a novel GRPR-targeted radioligand [111In]In-AMTG-DA1 containing an ALB moiety to improve tumor accumulation. [111In]In-AMTG-DA1 showed marked binding ability to albumin, high affinity for GRPR, and high-level stability in vitro. In biodistribution studies, the tumor accumulation of [111In]In-AMTG-DA1 was much higher than that of the control ligand without an ALB moiety. The introduction of ALB increased the tumor area under the curve (AUC) value of [111In]In-AMTG-DA1 by 3.5 times. In a single-photon emission computed tomography (SPECT) study, [111In]In-AMTG-DA1 visualized a GRPR-expressing tumor clearly at 24 h postinjection. Our findings suggest the favorable pharmacokinetics of [111In]In-AMTG-DA1 as a GRPR-targeted radioligand exhibiting a high-level accumulation in tumors.
Collapse
Affiliation(s)
- Shohei Tsuchihashi
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuma Nakashima
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
2
|
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] [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
|
3
|
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
|
4
|
Zhang H, Qi L, Cai Y, Gao X. Gastrin-releasing peptide receptor (GRPR) as a novel biomarker and therapeutic target in prostate cancer. Ann Med 2024; 56:2320301. [PMID: 38442298 PMCID: PMC10916925 DOI: 10.1080/07853890.2024.2320301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
Aim: This comprehensive review aims to explore the potential applications of Gastrin-releasing peptide receptor (GRPR) in the diagnosis and treatment of prostate cancer. Additionally, the study investigates the role of GRPR in prognostic assessment for individuals afflicted with prostate cancer.Methods: The review encompasses a thorough examination of existing literature and research studies related to the upregulation of GRPR in various tumor types, with a specific focus on prostate. The review also evaluates the utility of GRPR as a molecular target in prostate cancer research, comparing its significance to the well-established Prostate-specific membrane antigen (PSMA). The integration of radionuclide-targeted therapy with GRPR antagonists is explored as an innovative therapeutic approach for individuals with prostate cancer.Results: Research findings suggest that GRPR serves as a promising molecular target for visualizing low-grade prostate cancer. Furthermore, it is demonstrated to complement the detection of lesions that may be negative for PSMA. The integration of radionuclide-targeted therapy with GRPR antagonists presents a novel therapeutic paradigm, offering potential benefits for individuals undergoing treatment for prostate cancer.Conclusions: In conclusion, this review highlights the emerging role of GRPR in prostate cancer diagnosis and treatment. Moreover, the integration of radionuclide-targeted therapy with GRPR antagonists introduces an innovative therapeutic approach that holds promise for improving outcomes in individuals dealing with prostate cancer. The potential prognostic value of GRPR in assessing the disease's progression adds another dimension to its clinical significance in the realm of urology.
Collapse
Affiliation(s)
- Honghu Zhang
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, P. R. China
| | - Lin Qi
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, P. R. China
| | - Yi Cai
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, P. R. China
| | - Xiaomei Gao
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, P. R. China
| |
Collapse
|
5
|
Dong F, Hao L, Wang L, Huang Y. Clickable nanozyme enhances precise colonization of probiotics for ameliorating inflammatory bowel disease. J Control Release 2024; 373:749-765. [PMID: 39084465 DOI: 10.1016/j.jconrel.2024.07.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Convincing evidence suggests that aberrant gut microbiota changes play a critical role in the progression and pathogenesis of inflammatory bowel disease (IBD). Probiotic therapeutic interventions targeting the microbiota may provide alternative avenues to treat IBD, but currently available probiotics often suffer from low intestinal colonization and limited targeting capability. Here, we developed azido (N3)-modified Prussian blue nanozyme (PB@N3) spatio-temporal guidance enhances the targeted colonization of probiotics to alleviate intestinal inflammation. First, clickable PB@N3 targets intestinal inflammation, simultaneously, it scavenges reactive oxygen species (ROS). Subsequently, utilizing "click" chemistry to spatio-temporally guide targeted colonization of dibenzocyclooctyne (DBCO)-modified Lactobacillus reuteri DSM 17938 (LR@DBCO). The "click" reaction between PB@N3 and LR@DBCO has excellent specificity and efficacy both in vivo and in vitro. Despite the complex physiological environment of IBD, "click" reaction can prolong the retention time of probiotics in the intestine. Dextran sulfate sodium (DSS)-induced colitis mice model, demonstrates that the combination of PB@N3 and LR@DBCO effectively mitigates levels of ROS, enhances the colonization of probiotics, modulates intestinal flora composition and function, regulates immune profiles, restores intestinal barrier function, and alleviates intestinal inflammation. Hence, PB@N3 spatio-temporal guidance enhances targeted colonization of LR@DBCO provides a promising medical treatment strategy for IBD.
Collapse
Affiliation(s)
- Fang Dong
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Liangwen Hao
- The Institute for Biomedical Engineering and Nano Science School of Medicine, Tongji University, Shanghai 200072, China
| | - Lin Wang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ying Huang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
6
|
Yukawa H, Sato K, Baba Y. Theranostics applications of quantum dots in regenerative medicine, cancer medicine, and infectious diseases. Adv Drug Deliv Rev 2023; 200:114863. [PMID: 37156265 DOI: 10.1016/j.addr.2023.114863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023]
Abstract
Quantum dots (QDs) have attracted attention for their application and commercialization in all industrial fields, including communications, displays, and solar cells, due to their excellent optical properties based on the quantum size effect. In recent years, the development of QDs that do not contain cadmium which is toxic to cells and living organisms, has progressed, and they have attracted considerable attention in the bio-imaging field for targeting molecules and cells. Furthermore, recently, the need for diagnostics and treatment at the single molecule and single cell level in the medical field has been increasing, and the application of QDs in the medical field is also accelerating. Therefore, this paper outlines the frontiers of diagnostic and therapeutic applications (theranostics) of QDs, especially in advanced medical fields such as regenerative medicine, oncology, and infectious diseases.
Collapse
Affiliation(s)
- Hiroshi Yukawa
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya 466-8550, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya 466-8550, Japan; Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan; Department of Quantum Life Science, Graduate School of Science, Chiba University, Chiba 265-8522, Japan.
| | - Kazuhide Sato
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya 466-8550, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya 466-8550, Japan; Nagoya University Graduate School of Medicine, 65 Tsuruma, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya 466-8550, Japan; Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan.
| |
Collapse
|
7
|
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
|
8
|
Koustoulidou S, Handula M, de Ridder C, Stuurman D, Beekman S, de Jong M, Nonnekens J, Seimbille Y. Synthesis and Evaluation of Two Long-Acting SSTR2 Antagonists for Radionuclide Therapy of Neuroendocrine Tumors. Pharmaceuticals (Basel) 2022; 15:ph15091155. [PMID: 36145375 PMCID: PMC9503898 DOI: 10.3390/ph15091155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
Somatostatin receptor subtype 2 (SSTR2) has become an essential target for radionuclide therapy of neuroendocrine tumors (NETs). JR11 was introduced as a promising antagonist peptide to target SSTR2. However, due to its rapid blood clearance, a better pharmacokinetic profile is necessary for more effective treatment. Therefore, two JR11 analogs (8a and 8b), each carrying an albumin binding domain, were designed to prolong the blood residence time of JR11. Both compounds were labeled with lutetium-177 and evaluated via in vitro assays, followed by in vivo SPECT/CT imaging and ex vivo biodistribution studies. [177Lu]Lu-8a and [177Lu]Lu-8b were obtained with high radiochemical purity (>97%) and demonstrated excellent stability in PBS and mouse serum (>95%). [177Lu]Lu-8a showed better affinity towards human albumin compared to [177Lu]Lu-8b. Further, 8a and 8b exhibited binding affinities 30- and 48-fold lower, respectively, than that of the parent peptide JR11, along with high cell uptake and low internalization rate. SPECT/CT imaging verified high tumor accumulation for [177Lu]Lu-8a and [177Lu]Lu-JR11 at 4, 24, 48, and 72 h post-injection, but no tumor uptake was observed for [177Lu]Lu-8b. Ex vivo biodistribution studies revealed high and increasing tumor uptake for [177Lu]Lu-8a. However, its extended blood circulation led to an unfavorable biodistribution profile for radionuclide therapy.
Collapse
Affiliation(s)
- Sofia Koustoulidou
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Savanne Beekman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Julie Nonnekens
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
- Correspondence: ; Tel.: +31-10-703-8961
| |
Collapse
|
9
|
Value of 68Ga-labeled bombesin antagonist (RM2) in the detection of primary prostate cancer comparing with [ 18F]fluoromethylcholine PET-CT and multiparametric MRI-a phase I/II study. Eur Radiol 2022; 33:472-482. [PMID: 35864350 DOI: 10.1007/s00330-022-08982-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/12/2022] [Accepted: 06/12/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES The bombesin derivative RM2 is a GRPr antagonist with strong binding affinity to prostate cancer (PCa). In this study, the impact of [68Ga]Ga-RM2 positron emission tomography-computed tomography (PET-CT) for the detection of primary PCa was compared with that of [18F]FCH PET-CT and multiparametric magnetic resonance imaging (mpMRI). METHODS This phase I/II study was conducted in 30 biopsy-positive PCa subjects. The patients were stratified into high (10 patients), intermediate (10 patients), and low risk (10 patients) for extraglandular metastases as defined by National Comprehensive Cancer Network (NCCN) criteria (NCCN Clinical Practice Guidelines in Oncology, 2016). The prostate gland was classified in 12 anatomic segments for data analysis of the imaging modalities as well as histopathologic findings. The segment with the highest radiotracer uptake was defined as the "index lesion." All cases were scheduled to undergo prostatectomy with pelvic lymph node (LN) dissection in intermediate- and high-risk patients. Intraprostatic and pelvic nodal [68Ga]Ga-RM2 and [18F]FCH PET-CT findings were correlated with mpMRI and histopathologic results. RESULTS Of the 312 analyzed regions, 120 regions (4 to 8 lesions per patient) showed abnormal findings in the prostate gland. In a region-based analysis, overall sensitivity and specificity of [68Ga]Ga-RM2 PET-CT in the detection of primary tumor were 74% and 90%, respectively, while it was 60% and 80% for [18F]FCH PET-CT and 72% and 89% for mpMRI. Although the overall sensitivity of [68Ga]Ga-RM2 PET-CT was higher compared to that of [18F]FCH PET-CT and mpMRI, the statistical analysis showed only significant difference between [68Ga]Ga-RM2 PET-CT and [18F]FCH PET-CT in the intermediate-risk group (p = 0.01) and [68Ga]Ga-RM2 PET-CT and mpMRT in the high-risk group (p = 0.03). In the lesion-based analysis, there was no significant difference between SUVmax of [68Ga]Ga-RM2 and [18F]FCH PET-CT in the intraprostatic malignant lesions ([68Ga]Ga-RM2: mean SUVmax: 5.98 ± 4.13, median: 4.75; [18F]FCH: mean SUVmax: 6.08 ± 2.74, median: 5.5; p = 0.13). CONCLUSIONS [68Ga]Ga-RM2 showed promising PET tracer for the detection of intraprostatic PCa in a cohort of patients with different risk stratifications. However, significant differences were only found between [68Ga]Ga-RM2 PET-CT and [18F]FCH PET-CT in the intermediate-risk group and [68Ga]Ga-RM2 PET-CT and mpMRT in the high-risk group. In addition, GRP-R-based imaging seems to play a complementary role to choline-based imaging for full characterization of PCa extent and biopsy guidance in low- and intermediate-metastatic-risk PCa patients and has the potential to discriminate them from those at higher risks. KEY POINTS • [68Ga]Ga-RM2 is a promising PET tracer with a high detection rate for intraprostatic PCa especially in intermediate-risk prostate cancer patients. • GRPr-based imaging seems to play a complementary role to choline-based or PSMA-based PET/CT imaging in selected low- and intermediate-risk PCa patients for better characterization and eventually biopsy guidance of prostate cancer disease.
Collapse
|
10
|
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
|
11
|
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
|
12
|
Handula M, Verhoeven M, Chen KT, Haeck J, de Jong M, Dalm SU, Seimbille Y. Towards Complete Tumor Resection: Novel Dual-Modality Probes for Improved Image-Guided Surgery of GRPR-Expressing Prostate Cancer. Pharmaceutics 2022; 14:pharmaceutics14010195. [PMID: 35057090 PMCID: PMC8778164 DOI: 10.3390/pharmaceutics14010195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 02/01/2023] Open
Abstract
Nuclear and optical dual-modality probes can be of great assistance in prostate cancer localization, providing the means for both preoperative nuclear imaging and intraoperative surgical guidance. We developed a series of probes based on the backbone of the established GRPR-targeting radiotracer NeoB. The inverse electron demand of the Diels–Alder reaction was used to integrate the sulfo-cyanine 5 dye. Indium-111 radiolabeling, stability studies and a competition binding assay were carried out. Pilot biodistribution and imaging studies were performed in PC-3 tumor-bearing mice, using the best two dual-labeled probes. The dual-modality probes were radiolabeled with a high yield (>92%), were proven to be hydrophilic and demonstrated high stability in mouse serum (>94% intact labeled ligand at 4 h). The binding affinity for the GRPR was in the nanomolar range (21.9–118.7 nM). SPECT/CT images at 2 h p.i. clearly visualized the tumor xenograft and biodistribution studies, after scanning confirmed the high tumor uptake (8.47 ± 0.46%ID/g and 6.90 ± 0.81%ID/g for probe [111In]In-12 and [111In]In-15, respectively). Receptor specificity was illustrated with blocking studies, and co-localization of the radioactive and fluorescent signal was verified by ex vivo fluorescent imaging. Although optimal tumor-to-blood and tumor-to-kidney ratios might not yet have been reached due to the prolonged blood circulation, our probes are promising candidates for the preoperative and intraoperative visualization of GRPR-positive prostate cancer.
Collapse
Affiliation(s)
- Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Marjolein Verhoeven
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Kuo-Ting Chen
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien 974301, Taiwan;
| | - Joost Haeck
- AMIE Core Facility, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Simone U. Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.H.); (M.V.); (S.U.D.)
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
- Correspondence: ; Tel.: +31-10-703-8961
| |
Collapse
|
13
|
Li H, Kim Y, Jung H, Hyun JY, Shin I. Near-infrared (NIR) fluorescence-emitting small organic molecules for cancer imaging and therapy. Chem Soc Rev 2022; 51:8957-9008. [DOI: 10.1039/d2cs00722c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We discuss recent advances made in the development of NIR fluorescence-emitting small organic molecules for tumor imaging and therapy.
Collapse
Affiliation(s)
- Hui Li
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| | - Yujun Kim
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| | - Hyoje Jung
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea
| |
Collapse
|
14
|
Heing‐Becker I, Grötzinger C, Beindorff N, Prasad S, Erdmann S, Exner S, Haag R, Licha K. A Cyanine-Bridged Somatostatin Hybrid Probe for Multimodal SSTR2 Imaging in Vitro and in Vivo: Synthesis and Evaluation. Chembiochem 2021; 22:1307-1315. [PMID: 33238069 PMCID: PMC8048842 DOI: 10.1002/cbic.202000791] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/20/2022]
Abstract
Multimodal imaging probes have attracted the interest of ongoing research, for example, for the surgical removal of tumors. Modular synthesis approaches allow the construction of hybrid probes consisting of a radiotracer, a fluorophore and a targeting unit. We present the synthesis of a new asymmetric bifunctional cyanine dye that can be used as a structural and functional linker for the construction of such hybrid probes. 68 Ga-DOTATATE, a well-characterized radiopeptide targeting the overexpressed somatostatin receptor subtype 2 (SSTR2) in neuroendocrine tumors, was labeled with our cyanine dye, thus providing additional information along with the data obtained from the radiotracer. We tested the SSTR2-targeting and imaging properties of the resulting probe 68 Ga-DOTA-ICC-TATE in vitro and in a tumor xenograft mouse model. Despite the close proximity between dye and pharmacophore, we observed a high binding affinity towards SSTR2 as well as elevated uptake in SSTR2-overexpressing tumors in the positron emission tomography (PET) scan and histological examination.
Collapse
Affiliation(s)
- Isabelle Heing‐Becker
- Institut für Chemie und BiochemieFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Carsten Grötzinger
- Department of Hepatology and GastroenterologyCharité – Universitätsmedizin BerlinAugustenburger Platz 113353BerlinGermany
| | - Nicola Beindorff
- BERIC – Berlin Experimental Radionuclide Imaging CenterCharité – Universitätsmedizin BerlinAugustenburger Platz 113353BerlinGermany
| | - Sonal Prasad
- BERIC – Berlin Experimental Radionuclide Imaging CenterCharité – Universitätsmedizin BerlinAugustenburger Platz 113353BerlinGermany
- Department of Nuclear MedicineCharité – Universitätsmedizin BerlinAugustenburger Platz 113353BerlinGermany
| | - Sarah Erdmann
- Department of Hepatology and GastroenterologyCharité – Universitätsmedizin BerlinAugustenburger Platz 113353BerlinGermany
| | - Samantha Exner
- Department of Hepatology and GastroenterologyCharité – Universitätsmedizin BerlinAugustenburger Platz 113353BerlinGermany
| | - Rainer Haag
- Institut für Chemie und BiochemieFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Kai Licha
- Institut für Chemie und BiochemieFreie Universität BerlinTakustr. 314195BerlinGermany
| |
Collapse
|
15
|
Biodistribution and Radiation Dosimetric Analysis of [68Ga]Ga-RM2: A Potent GRPR Antagonist in Prostate Carcinoma Patients. RADIATION 2020. [DOI: 10.3390/radiation1010004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
[68Ga]Ga-RM2 is a promising innovative positron emission tomography (PET) tracer for patients with primary or metastatic prostate carcinoma. This study aims to analyze the biodistribution and radiation dosimetry of [68Ga]Ga-RM2 in five prostate cancer patients. The percentages of injected activity in the source organs and blood samples were determined. Bone marrow residence time was calculated using an indirect blood-based method. OLINDA/EXM version 2.0 (Hermes Medical Solutions, Stockholm, Sweden) was used to determine residence times, organ absorbed and effective doses. Physiological uptake was seen in kidneys, urinary bladder, pancreas, stomach, spleen and liver. Blood clearance was fast and followed by rapid clearance of activity from kidneys resulting in high activity concentrations in the urinary bladder. The urinary bladder wall was the most irradiated organ with highest mean organ absorbed dose (0.470 mSv/MBq) followed by pancreas (0.124 mSv/MBq), stomach wall (0.063 mSv/MBq), kidneys (0.049 mSv/MBq) and red marrow (0.010 mSv/MBq). The effective dose was found to be 0.038 mSv/MBq. Organ absorbed doses were found to be comparable to other gallium-68 labelled GRPR antagonists and lower than [68Ga]Ga-PSMA with the exception of the urinary bladder, pancreas and stomach wall. Remarkable interindividual differences were observed for the organ absorbed doses. Therefore, [68Ga]Ga-RM2 is a safe diagnostic agent with a significantly lower kidney dose but higher pancreas and urinary bladder doses as compared to [68Ga]Ga-PSMA.
Collapse
|
16
|
Hernandez Vargas S, Lin C, Voss J, Ghosh SC, Halperin DM, AghaAmiri S, Cao HST, Ikoma N, Uselmann AJ, Azhdarinia A. Development of a drug-device combination for fluorescence-guided surgery in neuroendocrine tumors. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200129R. [PMID: 33300316 PMCID: PMC7725236 DOI: 10.1117/1.jbo.25.12.126002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/06/2020] [Indexed: 05/13/2023]
Abstract
SIGNIFICANCE The use of cancer-targeted contrast agents in fluorescence-guided surgery (FGS) has the potential to improve intraoperative visualization of tumors and surgical margins. However, evaluation of their translational potential is challenging. AIM We examined the utility of a somatostatin receptor subtype-2 (SSTR2)-targeted fluorescent agent in combination with a benchtop near-infrared fluorescence (NIRF) imaging system to visualize mouse xenografts under conditions that simulate the clinical FGS workflow for open surgical procedures. APPROACH The dual-labeled somatostatin analog, Ga67-MMC(IR800)-TOC, was injected into mice (n = 24) implanted with SSTR2-expressing tumors and imaged with the customized OnLume NIRF imaging system (Madison, Wisconsin). In vivo and ex vivo imaging were performed under ambient light. The optimal dose (0.2, 0.5, and 2 nmol) and imaging time point (3, 24, 48, and 72 h) were determined using contrast-to-noise ratio (CNR) as the image quality parameter. Video captures of tumor resections were obtained to provide an FGS readout that is representative of clinical utility. Finally, a log-transformed linear regression model was fitted to assess congruence between fluorescence readouts and the underlying drug distribution. RESULTS The drug-device combination provided high in vivo and ex vivo contrast (CNRs > 3, except lung at 3 h) at all time points with the optimal dose of 2 nmol. The optimal imaging time point was 24-h post-injection, where CNRs > 6.5 were achieved in tissues of interest (i.e., pancreas, small intestine, stomach, and lung). Intraoperative FGS showed excellent utility for examination of the tumor cavity pre- and post-resection. The relationship between fluorescence readouts and gamma counts was linear and strongly correlated (n = 334, R2 = 0.71; r = 0.84; P < 0.0001). CONCLUSION The innovative OnLume NIRF imaging system enhanced the evaluation of Ga67-MMC(IR800)-TOC in tumor models. These components comprise a promising drug-device combination for FGS in patients with SSTR2-expressing tumors.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | | | - Julie Voss
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | - Sukhen C. Ghosh
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | - Daniel M. Halperin
- The University of Texas MD Anderson Cancer Center, Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, Houston, Texas, United States
| | - Solmaz AghaAmiri
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | - Hop S. Tran Cao
- The University of Texas MD Anderson Cancer Center, Department of Surgical Oncology, Division of Surgery, Houston, Texas, United States
| | - Naruhiko Ikoma
- The University of Texas MD Anderson Cancer Center, Department of Surgical Oncology, Division of Surgery, Houston, Texas, United States
| | | | - Ali Azhdarinia
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| |
Collapse
|
17
|
Klenner MA, Pascali G, Massi M, Fraser BH. Fluorine‐18 Radiolabelling and Photophysical Characteristics of Multimodal PET–Fluorescence Molecular Probes. Chemistry 2020; 27:861-876. [DOI: 10.1002/chem.202001402] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Mitchell A. Klenner
- Human Health and National Deuteration Facility (NDF) Australian Nuclear Science and Technology Organisation (ANSTO) New Illawarra Road Lucas Heights NSW 2234 Australia
- School of Molecular and Life Sciences Curtin University Kent Street Bentley WA 6102 Australia
| | - Giancarlo Pascali
- Human Health and National Deuteration Facility (NDF) Australian Nuclear Science and Technology Organisation (ANSTO) New Illawarra Road Lucas Heights NSW 2234 Australia
- Prince of Wales Hospital Barker St Randwick NSW 2031 Australia
- University of New South Wales Sydney (UNSW) Kensington NSW 2052 Australia
| | - Massimiliano Massi
- School of Molecular and Life Sciences Curtin University Kent Street Bentley WA 6102 Australia
| | - Benjamin H. Fraser
- Human Health and National Deuteration Facility (NDF) Australian Nuclear Science and Technology Organisation (ANSTO) New Illawarra Road Lucas Heights NSW 2234 Australia
| |
Collapse
|
18
|
Li Y, Zhou Y, Yue X, Dai Z. Cyanine Conjugate-Based Biomedical Imaging Probes. Adv Healthc Mater 2020; 9:e2001327. [PMID: 33000915 DOI: 10.1002/adhm.202001327] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/11/2020] [Indexed: 12/12/2022]
Abstract
Cyanine is a class of fluorescent dye with meritorious fluorescence properties and has motivated numerous researchers to explore its imaging capabilities by miscellaneous structural modification and functionalization strategies. The covalent conjugation with other functional molecules represents a distinctive design strategy and has shown immense potential in both basic and clinical research. This review article summarizes recent achievements in cyanine conjugate-based probes for biomedical imaging. Particular attention is paid to the conjugation with targeting warheads and other contrast agents for targeted fluorescence imaging and multimodal imaging, respectively. Additionally, their clinical potential in cancer diagnostics is highlighted and some concurrent impediments for clinical translation are discussed.
Collapse
Affiliation(s)
- Yang Li
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| | - Yiming Zhou
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| | - Xiuli Yue
- School of Environment Harbin Institute of Technology Harbin 150090 China
| | - Zhifei Dai
- Department of Biomedical Engineering College of Engineering Peking University Beijing 100871 China
| |
Collapse
|
19
|
Wang W, Wu K, Vellaisamy K, Leung C, Ma D. Peptide‐Conjugated Long‐Lived Theranostic Imaging for Targeting GRPr in Cancer and Immune Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202007920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Wanhe Wang
- Department of Chemistry Hong Kong Baptist University Kowloon, Hong Kong SAR 999077 China
| | - Ke‐Jia Wu
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine University of Macau Taipa, Macau SAR 999078 China
| | - Kasipandi Vellaisamy
- Department of Chemistry Hong Kong Baptist University Kowloon, Hong Kong SAR 999077 China
| | - Chung‐Hang Leung
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine University of Macau Taipa, Macau SAR 999078 China
| | - Dik‐Lung Ma
- Department of Chemistry Hong Kong Baptist University Kowloon, Hong Kong SAR 999077 China
| |
Collapse
|
20
|
Wang W, Wu K, Vellaisamy K, Leung C, Ma D. Peptide‐Conjugated Long‐Lived Theranostic Imaging for Targeting GRPr in Cancer and Immune Cells. Angew Chem Int Ed Engl 2020; 59:17897-17902. [DOI: 10.1002/anie.202007920] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Wanhe Wang
- Department of Chemistry Hong Kong Baptist University Kowloon, Hong Kong SAR 999077 China
| | - Ke‐Jia Wu
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine University of Macau Taipa, Macau SAR 999078 China
| | - Kasipandi Vellaisamy
- Department of Chemistry Hong Kong Baptist University Kowloon, Hong Kong SAR 999077 China
| | - Chung‐Hang Leung
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine University of Macau Taipa, Macau SAR 999078 China
| | - Dik‐Lung Ma
- Department of Chemistry Hong Kong Baptist University Kowloon, Hong Kong SAR 999077 China
| |
Collapse
|
21
|
Andrews JPM, Portal C, Walton T, Macaskill MG, Hadoke PWF, Alcaide Corral C, Lucatelli C, Wilson S, Wilson I, MacNaught G, Dweck MR, Newby DE, Tavares AAS. Non-invasive in vivo imaging of acute thrombosis: development of a novel factor XIIIa radiotracer. Eur Heart J Cardiovasc Imaging 2020; 21:673-682. [PMID: 31408105 PMCID: PMC7237957 DOI: 10.1093/ehjci/jez207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/01/2019] [Accepted: 08/07/2019] [Indexed: 11/12/2022] Open
Abstract
AIMS Cardiovascular thrombosis is responsible a quarter of deaths annually worldwide. Current imaging methods for cardiovascular thrombosis focus on anatomical identification of thrombus but cannot determine thrombus age or activity. Molecular imaging techniques hold promise for identification and quantification of thrombosis in vivo. Our objective was to assess a novel optical and positron-emitting probe targeting Factor XIIIa (ENC2015) as biomarker of active thrombus formation. METHODS AND RESULTS Optical and positron-emitting ENC2015 probes were assessed ex vivo using blood drawn from human volunteers and passed through perfusion chambers containing denuded porcine aorta as a model of arterial injury. Specificity of ENC2015 was established with co-infusion of a factor XIIIa inhibitor. In vivo18F-ENC2015 biodistribution, kinetics, radiometabolism, and thrombus binding were characterized in rats. Both Cy5 and fluorine-18 labelled ENC2015 rapidly and specifically bound to thrombi. Thrombus uptake was inhibited by a factor XIIIa inhibitor. 18F-ENC2015 remained unmetabolized over 8 h when incubated in ex vivo human blood. In vivo, 42% of parent radiotracer remained in blood 60 min post-administration. Biodistribution studies demonstrated rapid clearance from tissues with elimination via the urinary system. In vivo,18F-ENC2015 uptake was markedly increased in the thrombosed carotid artery compared to the contralateral patent artery (mean standard uptake value ratio of 2.40 vs. 0.74, P < 0.0001). CONCLUSION ENC2015 rapidly and selectively binds to acute thrombus in both an ex vivo human translational model and an in vivo rodent model of arterial thrombosis. This probe holds promise for the non-invasive identification of thrombus formation in cardiovascular disease.
Collapse
Affiliation(s)
- Jack P M Andrews
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK, Corresponding author. Tel: +44 (77) 6688 5010; Fax: +131 242 6379. E-mail:
| | - Christophe Portal
- Edinburgh Molecular Imaging Ltd., 9 Little France Road, Edinburgh, UK
| | - Tashfeen Walton
- Edinburgh Imaging facility QMRI, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Mark G Macaskill
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| | - Patrick W F Hadoke
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| | - Carlos Alcaide Corral
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| | - Christophe Lucatelli
- Edinburgh Imaging facility QMRI, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Simon Wilson
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| | - Ian Wilson
- ImaginAb, Inc. U.S. 43 Hindry Avenue, Suite D, Inglewood, California, USA
| | - Gillian MacNaught
- Edinburgh Imaging facility QMRI, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| | - David E Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| | - Adriana A S Tavares
- BHF Centre for Cardiovascular Science, University of Edinburgh, 49 Little France Crescent, Edinburgh, UK
| |
Collapse
|
22
|
Real-time, functional intra-operative localization of rat cavernous nerve network using near-infrared cyanine voltage-sensitive dye imaging. Sci Rep 2020; 10:6618. [PMID: 32313132 PMCID: PMC7171155 DOI: 10.1038/s41598-020-63588-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/27/2020] [Indexed: 12/02/2022] Open
Abstract
Despite current progress achieved in the surgical technique of radical prostatectomy, post-operative complications such as erectile dysfunction and urinary incontinence persist at high incidence rates. In this paper, we present a methodology for functional intra-operative localization of the cavernous nerve (CN) network for nerve-sparing radical prostatectomy using near-infrared cyanine voltage-sensitive dye (VSD) imaging, which visualizes membrane potential variations in the CN and its branches (CNB) in real time. As a proof-of-concept experiment, we demonstrate a functioning complex nerve network in response to electrical stimulation of the CN, which was clearly differentiated from surrounding tissues in an in vivo rat prostate model. Stimulation of an erection was confirmed by correlative intracavernosal pressure (ICP) monitoring. Within 10 minutes, we performed trans-fascial staining of the CN by direct VSD administration. Our findings suggest the applicability of VSD imaging for real-time, functional imaging guidance during nerve-sparing radical prostatectomy.
Collapse
|
23
|
Wang Y, Weng J, Lin J, Ye D, Zhang Y. NIR Scaffold Bearing Three Handles for Biocompatible Sequential Click Installation of Multiple Functional Arms. J Am Chem Soc 2020; 142:2787-2794. [PMID: 31944682 DOI: 10.1021/jacs.9b10467] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Near-infrared (NIR) probes are ideal for fluorescence labeling and imaging of biological targets in living animals. However, the instability of common NIR dyes hampers the construction of NIR probes bearing multiple functional components such as biomolecules for specific targeting and imaging reagents for multimodality imaging. To overcome these limitations, we designed a novel NIR scaffold bearing two terminal alkynes as clickable handles and a chloride on the heptamethine backbone that allows nucleophilic substitution with an azide to generate the third clickable handle. This unique scaffold allows for facile installation of multiple functional arms for the construction of multifunctional NIR probes. Various biomacromolecules or imaging reagents can be introduced to the NIR scaffold by sequential one-pot click reactions under biocompatible conditions. The preclickable handle chloride on the NIR backbone does not interfere with the initial click reactions, and it can be easily transformed into an azide for a following click reaction. On the basis of this unique NIR scaffold, we developed a highly efficient method to construct diverse NIR probes containing multiple functional biomolecules including peptides, antibodies, nucleic acids, and NIR/PET (positron emission tomography) dual-modality imaging probes bearing tumor-targeting groups. NIR imaging or multimodality imaging using these probes was performed on live cells or tumor models on living mice.
Collapse
Affiliation(s)
- Yuqi Wang
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC) , Nanjing University , Nanjing 210023 , China
| | - Jianhui Weng
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC) , Nanjing University , Nanjing 210023 , China
| | - Jianguo Lin
- Key Laboratory of Nuclear Medicine of Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine , Jiangsu Institute of Nuclear Medicine , Wuxi 214063 , China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC) , Nanjing University , Nanjing 210023 , China
| | - Yan Zhang
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC) , Nanjing University , Nanjing 210023 , China
| |
Collapse
|
24
|
Chen KT, Nieuwenhuizen J, Handula M, Seimbille Y. A novel clickable MSAP agent for dual fluorescence/nuclear labeling of biovectors. Org Biomol Chem 2020; 18:6134-6139. [DOI: 10.1039/d0ob01222j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Simple and efficient synthesis of dual-modality imaging agents for preoperative surgical planning and intraoperative surgical guidance.
Collapse
Affiliation(s)
- Kuo-Ting Chen
- Erasmus MC
- University Medical Center Rotterdam
- Department of Radiology and Nuclear Medicine
- Rotterdam
- The Netherlands
| | - Jim Nieuwenhuizen
- Erasmus MC
- University Medical Center Rotterdam
- Department of Radiology and Nuclear Medicine
- Rotterdam
- The Netherlands
| | - Maryana Handula
- Erasmus MC
- University Medical Center Rotterdam
- Department of Radiology and Nuclear Medicine
- Rotterdam
- The Netherlands
| | - Yann Seimbille
- Erasmus MC
- University Medical Center Rotterdam
- Department of Radiology and Nuclear Medicine
- Rotterdam
- The Netherlands
| |
Collapse
|
25
|
Chen F, Madajewski B, Ma K, Karassawa Zanoni D, Stambuk H, Turker MZ, Monette S, Zhang L, Yoo B, Chen P, Meester RJC, de Jonge S, Montero P, Phillips E, Quinn TP, Gönen M, Sequeira S, de Stanchina E, Zanzonico P, Wiesner U, Patel SG, Bradbury MS. Molecular phenotyping and image-guided surgical treatment of melanoma using spectrally distinct ultrasmall core-shell silica nanoparticles. SCIENCE ADVANCES 2019; 5:eaax5208. [PMID: 31840066 PMCID: PMC6892625 DOI: 10.1126/sciadv.aax5208] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/25/2019] [Indexed: 05/22/2023]
Abstract
Accurate detection and quantification of metastases in regional lymph nodes remain a vital prognostic predictor for cancer staging and clinical outcomes. As intratumoral heterogeneity poses a major hurdle to effective treatment planning, more reliable image-guided, cancer-targeted optical multiplexing tools are critically needed in the operative suite. For sentinel lymph node mapping indications, accurately interrogating distinct molecular signatures on cancer cells in vivo with differential levels of sensitivity and specificity remains largely unexplored. To address these challenges and demonstrate sensitivity to detecting micrometastases, we developed batches of spectrally distinct 6-nm near-infrared fluorescent core-shell silica nanoparticles, each batch surface-functionalized with different melanoma targeting ligands. Along with PET imaging, particles accurately detected and molecularly phenotyped cancerous nodes in a spontaneous melanoma miniswine model using image-guided multiplexing tools. Information afforded from these tools offers the potential to not only improve the accuracy of targeted disease removal and patient safety, but to transform surgical decision-making for oncological patients.
Collapse
Affiliation(s)
- Feng Chen
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Brian Madajewski
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Kai Ma
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Daniella Karassawa Zanoni
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hilda Stambuk
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Melik Z. Turker
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sébastien Monette
- Laboratory of Comparative Pathology, Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Li Zhang
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Barney Yoo
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Peiming Chen
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | | | - Sander de Jonge
- Quest Medical Imaging B.V., NL-1775PW, Middenmeer, Netherlands
| | - Pablo Montero
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Evan Phillips
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Thomas P. Quinn
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
- Harry S Truman Veterans’ Hospital, Columbia, MO 65201, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Sonia Sequeira
- Research and Technology Management, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Elisa de Stanchina
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ulrich Wiesner
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Snehal G. Patel
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michelle S. Bradbury
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| |
Collapse
|
26
|
Zhang R, Xu Y, Zhang Y, Kim HS, Sharma A, Gao J, Yang G, Kim JS, Sun Y. Rational design of a multifunctional molecular dye for dual-modal NIR-II/photoacoustic imaging and photothermal therapy. Chem Sci 2019; 10:8348-8353. [PMID: 31803412 PMCID: PMC6839587 DOI: 10.1039/c9sc03504d] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
Small-molecule based multifunctional probes play significant roles in biomedical science and possess high clinical translational ability. However, the preparation of these promising probes without complicated synthetic procedures remains a challenging task. Herein, we rationally designed a high-performance DD-A-DD scaffold molecular dye (SYL) with an intrinsic multifunctional ability and then incorporated it into DSPE-mPEG5000 to facilely construct biocompatible NIR-II fluorescent and photoacoustic (PA) dual-modal theranostic nanoprobes (SYL NPs) (∼120 nm). In vivo studies confirmed that SYL NPs exhibited bright NIR-II fluorescence and PA signals in the tumor region with a promising signal to background ratio (S/B). Meanwhile, SYL NPs demonstrated significantly inhibited tumor growth under laser irradiation with no noticeable side effects. These promising results highlighted SYL NPs as a potential theranostic platform for cancer diagnosis (NIR-II region) and therapy.
Collapse
Affiliation(s)
- Ruiping Zhang
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Center of Chemical Biology , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
- Affiliated Da Yi Hospital of Shanxi Medical University , Taiyuan 020001 , China
| | - Yuling Xu
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Center of Chemical Biology , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
| | - Yi Zhang
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Center of Chemical Biology , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
| | - Hyeong Seok Kim
- Department of Chemistry , Korea University , Seoul 02841 , Korea
| | - Amit Sharma
- Department of Chemistry , Korea University , Seoul 02841 , Korea
| | - Jing Gao
- Jiangsu Key Laboratory of Medical Optics , Suzhou Institute of Biomedical Engineering and Technology , Chinese Academy of Sciences , Suzhou215163 , China
| | - Guangfu Yang
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Center of Chemical Biology , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
| | - Jong Seung Kim
- Department of Chemistry , Korea University , Seoul 02841 , Korea
| | - Yao Sun
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Center of Chemical Biology , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
| |
Collapse
|
27
|
Debie P, Hernot S. Emerging Fluorescent Molecular Tracers to Guide Intra-Operative Surgical Decision-Making. Front Pharmacol 2019; 10:510. [PMID: 31139085 PMCID: PMC6527780 DOI: 10.3389/fphar.2019.00510] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Fluorescence imaging is an emerging technology that can provide real-time information about the operating field during cancer surgery. Non-specific fluorescent agents, used for the assessment of blood flow and sentinel lymph node detection, have so far dominated this field. However, over the last decade, several clinical studies have demonstrated the great potential of targeted fluorescent tracers to visualize tumor lesions in a more specific way. This has led to an exponential growth in the development of novel molecular fluorescent contrast agents. In this review, the design of fluorescent molecular tracers will be discussed, with particular attention for agents and approaches that are of interest for clinical translation.
Collapse
Affiliation(s)
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
28
|
Simons BW, Turtle NF, Ulmert DH, Abou DS, Thorek DLJ. PSMA expression in the Hi-Myc model; extended utility of a representative model of prostate adenocarcinoma for biological insight and as a drug discovery tool. Prostate 2019; 79:678-685. [PMID: 30656716 PMCID: PMC6519119 DOI: 10.1002/pros.23770] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022]
Abstract
Prostate-specific membrane antigen (PSMA), also known as glutamate carboxypeptidase II (GCPII), is highly overexpressed in primary and metastatic prostate cancer (PCa). This has led to the development of radiopharmaceuticals for targeted imaging and therapy under current clinical evaluation. Despite this progress, the exact biological role of the protein in prostate cancer development and progression has not been fully elucidated. This is in part because the human PSMA and mouse PSMA (mPSMA) have different patterns of anatomical expression which confound study in the most widely utilized model organisms. Most notably, mPSMA is not expressed in the healthy murine prostate. Here, we reveal that mPSMA is highly upregulated in the prostate adenocarcinoma of the spontaneous Hi-Myc mouse model, a highly accurate and well characterized mouse model of prostate cancer development. Antibody detection and molecular imaging tools are used to confirm that mPSMA is expressed from early prostatic intraepithelial neoplasia (PIN) through adenocarcinoma.
Collapse
Affiliation(s)
- Brian W. Simons
- Center for Comparative MedicineBaylor College of MedicineHoustonTexas
| | - Norman F. Turtle
- Radiological Chemistry Imaging LaboratoryMallinckrodt Institute of RadiologyWashington University in St. LouisSt. LouisMissouri
| | - David H. Ulmert
- Johnsson Comprehensive Cancer CenterUniversity of CaliforniaLos AngelesCalifornia
- Department of Molecular and Medical PharmacologyUniversity of California Los AngelesLos AngelesCalifornia
| | - Diane S. Abou
- Radiological Chemistry Imaging LaboratoryMallinckrodt Institute of RadiologyWashington University in St. LouisSt. LouisMissouri
- Radiology Cyclotron Facility, Mallinckrodt Institute of RadiologyWashington University in St. LouisSt. Louis,Missouri
| | - Daniel L. J. Thorek
- Radiological Chemistry Imaging LaboratoryMallinckrodt Institute of RadiologyWashington University in St. LouisSt. LouisMissouri
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouri
| |
Collapse
|
29
|
Pagoto A, Garello F, Marini GM, Tripepi M, Arena F, Bardini P, Stefania R, Lanzardo S, Valbusa G, Porpiglia F, Manfredi M, Aime S, Terreno E. Novel Gastrin-Releasing Peptide Receptor Targeted Near-Infrared Fluorescence Dye for Image-Guided Surgery of Prostate Cancer. Mol Imaging Biol 2019; 22:85-93. [DOI: 10.1007/s11307-019-01354-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
30
|
Zettlitz KA, Waldmann CM, Tsai WTK, Tavaré R, Collins J, Murphy JM, Wu AM. A Dual-Modality Linker Enables Site-Specific Conjugation of Antibody Fragments for 18F-Immuno-PET and Fluorescence Imaging. J Nucl Med 2019; 60:1467-1473. [PMID: 30877181 DOI: 10.2967/jnumed.118.223560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/06/2019] [Indexed: 12/30/2022] Open
Abstract
Antibody-based dual-modality (PET/fluorescence) imaging enables both presurgery antigen-specific immuno-PET for noninvasive whole-body evaluation and intraoperative fluorescence for visualization of superficial tissue layers for image-guided surgery. Methods: We developed a universal dual-modality linker (DML) that facilitates site-specific conjugation to a cysteine residue-bearing antibody fragment, introduction of a commercially available fluorescent dye (via an amine-reactive prosthetic group), and rapid and efficient radiolabeling via click chemistry with 18F-labeled trans-cyclooctene (18F-TCO). To generate a dual-modality antibody fragment-based imaging agent, the DML was labeled with the far-red dye sulfonate cyanine 5 (sCy5), site-specifically conjugated to the C-terminal cysteine of the anti-prostate stem cell antigen (PSCA) cys-diabody A2, and subsequently radiolabeled by click chemistry with 18F-TCO. The new imaging probe was evaluated in a human PSCA-positive prostate cancer xenograft model by sequential immuno-PET and optical imaging. Uptake in target tissues was confirmed by ex vivo biodistribution. Results: We successfully synthesized a DML for conjugation of a fluorescent dye and 18F. The anti-PSCA cys-diabody A2 was site-specifically conjugated with either DML or sCy5 and radiolabeled via click chemistry with 18F-TCO. Immuno-PET imaging confirmed in vivo antigen-specific targeting of prostate cancer xenografts as early as 1 h after injection. Rapid renal clearance of the 50-kDa antibody fragment enables same-day imaging. Optical imaging showed antigen-specific fluorescent signal in PSCA-positive xenografts and high contrast to surrounding tissue and PSCA-negative xenografts. Conclusion: The DML enables site-specific conjugation away from the antigen-binding site of antibody fragments, with a controlled linker-to-protein ratio, and combines signaling moieties for 2 imaging systems into 1 molecule. Dual-modality imaging could provide both noninvasive whole-body imaging with organ-level biodistribution and fluorescence image-guided identification of tumor margins during surgery.
Collapse
Affiliation(s)
- Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Christopher M Waldmann
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Wen-Ting K Tsai
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Richard Tavaré
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jeffrey Collins
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jennifer M Murphy
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
31
|
Canovas C, Moreau M, Vrigneaud JM, Bellaye PS, Collin B, Denat F, Goncalves V. Modular Assembly of Multimodal Imaging Agents through an Inverse Electron Demand Diels-Alder Reaction. Bioconjug Chem 2019; 30:888-897. [PMID: 30742423 DOI: 10.1021/acs.bioconjchem.9b00017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The combination of two imaging probes on the same biomolecule gives access to targeted bimodal imaging agents that can provide more accurate diagnosis, complementary information, or that may be used in different applications, such as nuclear imaging and fluorescence guided surgery. In this study, we demonstrate that dichlorotetrazine, a small, commercially available compound, can be used as a modular platform to easily assemble various imaging probes. Doubly labeled tetrazines can then be conjugated to a protein through a biorthogonal IEDDA reaction. A series of difunctionalized tetrazine compounds containing various chelating agents and fluorescent dyes was synthesized. As a proof of concept, one of these bimodal probes was conjugated to trastuzumab, previously modified with a constrained alkyne group, and the resulting dual-labeled antibody was evaluated in a mouse model, bearing a HER2-positive tumor. A significant uptake into tumor tissues was observed in vivo, by both SPECT-CT and fluorescence imaging, and confirmed ex vivo in biodistribution studies.
Collapse
Affiliation(s)
- Coline Canovas
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR6302, CNRS , Université Bourgogne Franche-Comté , 9 avenue Alain Savary , 21000 , Dijon , France
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR6302, CNRS , Université Bourgogne Franche-Comté , 9 avenue Alain Savary , 21000 , Dijon , France
| | - Jean-Marc Vrigneaud
- Georges-François LECLERC Cancer Center - UNICANCER , 1 rue Pr Marion , 21079 , Dijon , France
| | - Pierre-Simon Bellaye
- Georges-François LECLERC Cancer Center - UNICANCER , 1 rue Pr Marion , 21079 , Dijon , France
| | - Bertrand Collin
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR6302, CNRS , Université Bourgogne Franche-Comté , 9 avenue Alain Savary , 21000 , Dijon , France.,Georges-François LECLERC Cancer Center - UNICANCER , 1 rue Pr Marion , 21079 , Dijon , France
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR6302, CNRS , Université Bourgogne Franche-Comté , 9 avenue Alain Savary , 21000 , Dijon , France
| | - Victor Goncalves
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR6302, CNRS , Université Bourgogne Franche-Comté , 9 avenue Alain Savary , 21000 , Dijon , France
| |
Collapse
|
32
|
Hernandez Vargas S, Ghosh SC, Azhdarinia A. New Developments in Dual-Labeled Molecular Imaging Agents. J Nucl Med 2019; 60:459-465. [PMID: 30733318 DOI: 10.2967/jnumed.118.213488] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
Intraoperative detection of tumors has had a profound impact on how cancer surgery is performed and addresses critical unmet needs in surgical oncology. Tumor deposits, margins, and residual cancer can be imaged through the use of fluorescent contrast agents during surgical procedures to complement visual and tactile guidance. The combination of fluorescent and nuclear contrast into a multimodality agent builds on these capabilities by adding quantitative, noninvasive nuclear imaging capabilities to intraoperative imaging. This review focuses on new strategies for the development and evaluation of targeted dual-labeled molecular imaging agents while highlighting the successful first-in-human application of this technique.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sukhen C Ghosh
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Ali Azhdarinia
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
33
|
Tsai WK, Zettlitz KA, Tavaré R, Kobayashi N, Reiter RE, Wu AM. Dual-Modality ImmunoPET/Fluorescence Imaging of Prostate Cancer with an Anti-PSCA Cys-Minibody. Am J Cancer Res 2018; 8:5903-5914. [PMID: 30613270 PMCID: PMC6299441 DOI: 10.7150/thno.27679] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/04/2018] [Indexed: 01/01/2023] Open
Abstract
Inadequate diagnostic methods for prostate cancer lead to over- and undertreatment, and the inability to intraoperatively visualize positive margins may limit the success of surgical resection. Prostate cancer visualization could be improved by combining the complementary modalities of immuno-positron emission tomography (immunoPET) for preoperative disease detection, and fluorescence imaging-guided surgery (FIGS) for real-time intraoperative tumor margin identification. Here, we report on the evaluation of dual-labeled humanized anti-prostate stem cell antigen (PSCA) cys-minibody (A11 cMb) for immunoPET/fluorescence imaging in subcutaneous and orthotopic prostate cancer models. Methods: A11 cMb was site-specifically conjugated with the near-infrared fluorophore Cy5.5 and radiolabeled with 124I or 89Zr. 124I-A11 cMb-Cy5.5 was used for successive immunoPET/fluorescence imaging of prostate cancer xenografts expressing high or moderate levels of PSCA (22Rv1-PSCA and PC3-PSCA). 89Zr-A11 cMb-Cy5.5 dual-modality imaging was evaluated in an orthotopic model. Ex vivo biodistribution at 24 h was used to confirm the uptake values, and tumors were visualized by post-mortem fluorescence imaging. Results: A11 cMb-Cy5.5 retained low nanomolar affinity for PSCA-positive cells. Conjugation conditions were established (dye-to-protein ratio of 0.7:1) that did not affect the biodistribution, pharmacokinetics, or clearance of A11 cMb. ImmunoPET using dual-labeled 124I-A11 cMb-Cy5.5 showed specific targeting to both 22Rv1-PSCA and PC3-PSCA s.c. xenografts in nude mice. Ex vivo biodistribution confirmed specific uptake to PSCA-expressing tumors with 22Rv1-PSCA:22Rv1 and PC3-PSCA:PC3 ratios of 13:1 and 5.6:1, respectively. Consistent with the immunoPET, fluorescence imaging showed a strong signal from both 22Rv1-PSCA and PC3-PSCA tumors compared with non-PSCA expressing tumors. In an orthotopic model, 89Zr-A11 cMb-Cy5.5 immunoPET was able to detect intraprostatically implanted 22Rv1-PSCA cells. Importantly, fluorescence imaging clearly distinguished the prostate tumor from surrounding seminal vesicles. Conclusion: Dual-labeled A11 cMb specifically visualized PSCA-positive tumor by successive immunoPET/fluorescence, which can potentially be translated for preoperative whole-body prostate cancer detection and intraoperative surgical guidance in patients.
Collapse
|
34
|
Xu H, Bandari RP, Lee L, Li R, Yu P, Smith CJ, Ma L. Design, Synthesis, and in Vitro and in Vivo Evaluation of High Affinity and Specificity Near-Infrared Fluorescent Bombesin Antagonists for Tumor Imaging. J Med Chem 2018; 61:7657-7670. [DOI: 10.1021/acs.jmedchem.8b00614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hang Xu
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Department of Chemical Engineering, University of Missouri, Columbia, Missouri 65211, United States
| | - Rajendra P. Bandari
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Li Lee
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri 65211, United States
| | - Ran Li
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Department of Stomatology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ping Yu
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri 65211, United States
| | - Charles J. Smith
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Lixin Ma
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|
35
|
Zhao J, Chen J, Ma S, Liu Q, Huang L, Chen X, Lou K, Wang W. Recent developments in multimodality fluorescence imaging probes. Acta Pharm Sin B 2018; 8:320-338. [PMID: 29881672 PMCID: PMC5989919 DOI: 10.1016/j.apsb.2018.03.010] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Multimodality optical imaging probes have emerged as powerful tools that improve detection sensitivity and accuracy, important in disease diagnosis and treatment. In this review, we focus on recent developments of optical fluorescence imaging (OFI) probe integration with other imaging modalities such as X-ray computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), single-photon emission computed tomography (SPECT), and photoacoustic imaging (PAI). The imaging technologies are briefly described in order to introduce the strengths and limitations of each techniques and the need for further multimodality optical imaging probe development. The emphasis of this account is placed on how design strategies are currently implemented to afford physicochemically and biologically compatible multimodality optical fluorescence imaging probes. We also present studies that overcame intrinsic disadvantages of each imaging technique by multimodality approach with improved detection sensitivity and accuracy.
Collapse
Affiliation(s)
- Jianhong Zhao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Junwei Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Shengnan Ma
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Qianqian Liu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Lixian Huang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Xiani Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyan Lou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131-0001, USA
| |
Collapse
|
36
|
Summer D, Grossrubatscher L, Petrik M, Michalcikova T, Novy Z, Rangger C, Klingler M, Haas H, Kaeopookum P, von Guggenberg E, Haubner R, Decristoforo C. Developing Targeted Hybrid Imaging Probes by Chelator Scaffolding. Bioconjug Chem 2017; 28:1722-1733. [PMID: 28462989 PMCID: PMC5481817 DOI: 10.1021/acs.bioconjchem.7b00182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Positron emission tomography (PET) as well as optical imaging (OI) with peptide receptor targeting probes have proven their value for oncological applications but also show restrictions depending on the clinical field of interest. Therefore, the combination of both methods, particularly in a single molecule, could improve versatility in clinical routine. This proof of principle study aims to show that a chelator, Fusarinine C (FSC), can be utilized as scaffold for novel dimeric dual-modality imaging agents. Two targeting vectors (a minigastrin analogue (MG11) targeting cholecystokinin-2 receptor overexpression (CCK2R) or integrin αVβ3 targeting cyclic pentapeptides (RGD)) and a near-infrared fluorophore (Sulfo-Cyanine7) were conjugated to FSC. The probes were efficiently labeled with gallium-68 and in vitro experiments including determination of logD, stability, protein binding, cell binding, internalization, and biodistribution studies as well as in vivo micro-PET/CT and optical imaging in U-87MG αVβ3- and A431-CCK2R expressing tumor xenografted mice were carried out. Novel bioconjugates showed high receptor affinity and highly specific targeting properties at both receptors. Ex vivo biodistribution and micro-PET/CT imaging studies revealed specific tumor uptake accompanied by slow blood clearance and retention in nontargeted tissues (spleen, liver, and kidneys) leading to visualization of tumors at early (30 to 120 min p.i.). Excellent contrast in corresponding optical imaging studies was achieved especially at delayed time points (24 to 72 h p.i.). Our findings show the proof of principle of chelator scaffolding for hybrid imaging agents and demonstrate FSC being a suitable bifunctional chelator for this approach. Improvements to fine-tune pharmacokinetics are needed to translate this into a clinical setting.
Collapse
Affiliation(s)
- Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Leo Grossrubatscher
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc , Hnevotinska 5, 779 00, Olomouc, Czech Republic
| | - Tereza Michalcikova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc , Hnevotinska 5, 779 00, Olomouc, Czech Republic
| | - Zbynek Novy
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc , Hnevotinska 5, 779 00, Olomouc, Czech Republic
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Maximilian Klingler
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Hubertus Haas
- Division of Molecular Biology/Biocenter, Medical University Innsbruck , Innrain 80-82, A-6020 Innsbruck, Austria
| | - Piriya Kaeopookum
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria.,Ministry of Science, Technology (MOST), Thailand Institute of Nuclear Technology (TINT) , Nakhonnayok 26120, Thailand
| | - Elisabeth von Guggenberg
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck , Anichstrasse 35, A-6020 Innsbruck, Austria
| |
Collapse
|
37
|
PET and PET/CT with radiolabeled choline in prostate cancer: a critical reappraisal of 20 years of clinical studies. Eur J Nucl Med Mol Imaging 2017; 44:1751-1776. [PMID: 28409220 DOI: 10.1007/s00259-017-3700-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/31/2017] [Indexed: 12/12/2022]
Abstract
We here aim to provide a comprehensive and critical review of the literature concerning the clinical applications of positron emission tomography/computed tomography (PET/CT) with radiolabeled choline in patients with prostate cancer (PCa). We will initially briefly summarize the historical context that brought to the synthesis of [11C]choline, which occurred exactly 20 years ago. We have arbitrarily grouped the clinical studies in three different periods, according to the year in which they were published and according to their relation with their applications in urology, radiotherapy and oncology. Studies at initial staging and, more extensively, studies in patients with biochemical failure, as well as factors predicting positive PET/CT will be reviewed. The capability of PET/CT with radiolabeled choline to provide prognostic information on PCa-specific survival will also be examined. The last sections will be devoted to the use of radiolabeled choline for monitoring the response to androgen deprivation therapy, radiotherapy, and chemotherapy. The accuracy and the limits of the technique will be discussed according to the information available from standard validation processes, including biopsy or histology. The clinical impact of the technique will be discussed on the basis of changes induced in the management of patients and in the evaluation of the response to therapy. Current indications to PET/CT, as officially endorsed by guidelines, or as routinely performed in the clinical practice will be illustrated. Emphasis will be made on methodological factors that might have influenced the results of the studies or their interpretation. Finally, we will briefly highlight the potential role of positron emission tomography/magnetic resonance and of new radiotracers for PCa imaging.
Collapse
|