1
|
Otani K, Zeniya T, Kawashima H, Moriguchi T, Nakano A, Han C, Murata S, Nishimura K, Koshino K, Yamahara K, Inubushi M, Iida H. Spatial and temporal tracking of multi-layered cells sheet using reporter gene imaging with human sodium iodide symporter: a preclinical study using a rat model of myocardial infarction. Eur J Nucl Med Mol Imaging 2024; 52:74-87. [PMID: 39207487 PMCID: PMC11599416 DOI: 10.1007/s00259-024-06889-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE This study aimed to evaluate a novel technique for cell tracking by visualising the activity of the human sodium/iodide symporter (hNIS) after transplantation of hNIS-expressing multilayered cell sheets in a rat model of chronic myocardial infarction. METHODS Triple-layered cell sheets were generated from mouse embryonic fibroblasts (MEFs) derived from mice overexpressing hNIS (hNIS-Tg). Myocardial infarction was induced by permanent ligation of the left anterior descending coronary artery in F344 athymic rats, and a triple-layered MEFs sheets were transplanted to the infarcted area two weeks after surgery. To validate the temporal tracking and kinetic analysis of the transplanted MEFs sheets, sequential cardiac single-photon emission computed tomography (SPECT) examinations with a 99mTcO4- injection were performed. The cell sheets generated using MEFs of wild-type mice (WT) served as controls. RESULTS A significantly higher amount of 99mTcO4- was taken into the hNIS-Tg MEFs than into WT MEFs (146.1 ± 30.9-fold). The obvious accumulation of 99mTcO4- was observed in agreement with the region where hNIS-Tg MEFs were transplanted, and these radioactivities peaked 40-60 min after 99mTcO4- administration. The volume of distribution of the hNIS-Tg MEF sheets declined gradually after transplantation, implying cellular malfunction and a loss in the number of transplanted cells. CONCLUSION The reporter gene imaging with hNIS enables the serial tracking and quantitative kinetic analysis of cell sheets transplanted to infarcted hearts.
Collapse
Affiliation(s)
- Kentaro Otani
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Tsutomu Zeniya
- Graduate School of Science and Technology, Hirosaki University, Aomori, Japan
| | - Hidekazu Kawashima
- Radioisotope Research Center, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tetsuaki Moriguchi
- Tandem Accelerator Complex (UTTAC), University of Tsukuba, Ibaraki, Japan
| | - Atsushi Nakano
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Chunlei Han
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Shunsuke Murata
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Kunihiro Nishimura
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Kazuhiro Koshino
- Department of Systems and Informatics, Hokkaido Information University, Hokkaido, Japan
| | - Kenichi Yamahara
- Laboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, Hyogo, Japan
| | - Masayuki Inubushi
- Division of Nuclear Medicine, Department of Radiology, Kawasaki Medical School, Okayama, Japan
| | - Hidehiro Iida
- Turku PET Centre, Turku University Hospital, Turku, Finland.
- Turku PET Centre, University of Turku and Turku University Hospital, Building 14, Kiinamyllynkatu 4-8, Turku, 20520, Finland.
| |
Collapse
|
2
|
Zhang K, Feng W, Mou Z, Zhang L, Ma M, Zhao Z, Liu X, Chen X, Li Z. Late-stage (radio)fluorination of alkyl phosphonates via electrophilic activation. Nat Commun 2024; 15:10338. [PMID: 39609392 PMCID: PMC11604993 DOI: 10.1038/s41467-024-54208-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024] Open
Abstract
Constructing organic fluorophosphines, vital drug skeletons, through the direct fluorination of readily available alkyl phosphonates has been impeded due to the intrinsic low electrophilicity of PV and the high bond energy of P═O bond. Here, alkyl phosphonates are electrophilically activated with triflic anhydride and N-heteroaromatic bases, enabling nucleophilic fluorination at room temperature to form fluorophosphines via reactive phosphine intermediates. This approach facilitates the late-stage (radio)fluorination of broad dialkyl and monoalkyl phosphonates. Monoalkyl phosphonates derived from targeted drugs, including cyclophosphamide, vortioxetine, and dihydrocholesterol, are effectively fluorinated, achieving notable yields of 47-71%. Radiofluorination of medically significant 18F-tracers and synthons are completed in radiochemical conversions (radio-TLC) of 51-88% and molar activities up to 251 ± 12 GBq/μmol (initial activity 11.2 GBq) within 10 min at room temperature. Utilizing a phosphonamidic fluoride building block (BFPA), [18F]BFPA-Flurpiridaz and [18F]BFPA-E[c(RGDyK)]2 demonstrate high-contrast target imaging, excellent pharmacokinetics, and negligible defluorination.
Collapse
Affiliation(s)
- Kaiqiang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China
| | - Wanru Feng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China
| | - Zhaobiao Mou
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China
| | - Lei Zhang
- Tianjin Engineering Technology Center of Chemical Wastewater Source Reduction and Recycling, School of Science, Tianjin Chengjian University, Tianjin, China
| | - Mengting Ma
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China
- Department of Nuclear Medicine, Xiang'an Hospital affiliated to Xiamen University, Xiamen, Fujian, China
| | - Zixiao Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China
| | - Xia Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, Singapore
| | - Zijing Li
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, China.
- Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, Fujian, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
3
|
Backhaus P, Pentlow KS, Ho AL, Mauguen A, Fagin JA, Pillarsetty NVK, Lyashchenko SK, Burnazi E, Ghossein RA, Chhabra S, Abusamra M, Larson SM, Schöder H, O'Donoghue J, Weber W, Grewal RK. [ 18F]TFB PET/CT misses intense [ 124I]iodine-avid metastases after redifferentiation therapy in metastatic thyroid cancer. EJNMMI Res 2024; 14:91. [PMID: 39377970 PMCID: PMC11461403 DOI: 10.1186/s13550-024-01138-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/12/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Fluorine 18-labelled tetrafluoroborate ([18F]TFB) is a substrate for the sodium/iodide symporter. In thyroid cancer, [18F]TFB-PET/CT may be an alternative to iodine imaging to evaluate the extent of disease, eligibility for radioiodine treatment, and success of redifferentiation therapies. We report the results of a pilot study to determine tumor uptake of [18F]TFB and compare its properties to [124I]IodinePET/CT in patients with metastatic thyroid cancer. METHODS Five patients were included in a prospective study. All patients received PET/CT 1 h after injection of 356 ± 12 MBq [18F]TFB and were given 230 ± 9 MBq [124I]Iodine orally on the same day, followed by PET/CT after 48 h. Before redifferentiation therapy, patients underwent an additional baseline [124I]Iodine PET/CT. Cases were analyzed by two board-certified specialists. Detection rates and Spearman correlation for [18F]TFB and [124I]Iodine were calculated. RESULTS Three patients had poorly differentiated thyroid cancer and received trametinib in a redifferentiation trial. Two patients had papillary thyroid cancer and did not receive redifferentiation therapy. Of the 33 lesions seen before/without redifferentiation therapy, 19 (58%) were visible on [18F]TFB and 30 (91%) on [124I]Iodine imaging. In the patients who underwent redifferentiation therapy, 48 lesions were newly seen on [124I]Iodine PET/CT with a median SUVmax of 3.3 (range, 0.4-285.0). All of these lesions were [18F]TFB-negative. CONCLUSION [18F]TFB failed to predict radioactive iodine uptake in patients with poorly differentiated thyroid cancer who underwent redifferentiation therapy with trametinib. It is unclear whether such discrepancies may also occur in other redifferentiation therapies or may even be encountered in redifferentiation-naïve thyroid cancer. TRIAL REGISTRATION NUMBER NCT03196518, registered on June 22, 2017.
Collapse
Affiliation(s)
- Philipp Backhaus
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Keith S Pentlow
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Audrey Mauguen
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James A Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Naga Vara Kishore Pillarsetty
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
| | - Serge K Lyashchenko
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
| | - Eva Burnazi
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Ronald A Ghossein
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shalini Chhabra
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
| | - Murad Abusamra
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
| | - Steven M Larson
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
| | - Heiko Schöder
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA
| | - Joseph O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Ravinder K Grewal
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, P.O. Box 77, New York, NY, 10065, USA.
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Calderoni L, Giovanella L, Fanti S. Endocrinology application of molecular imaging: current role of PET/CT. J Endocrinol Invest 2024; 47:2383-2396. [PMID: 38837101 PMCID: PMC11393017 DOI: 10.1007/s40618-024-02400-8] [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: 11/17/2023] [Accepted: 05/18/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND In recent years, nuclear medicine imaging methods have proven to be of paramount importance in a wide variety of diseases, particularly in oncology, where they are crucial for assessing the extent of disease when conventional methods fall short. Moreover, nuclear imaging modalities are able to better characterize lesions using target agents related to specific pathways (e.g. glucose metabolism, cellular proliferation, amino acid transport, lipid metabolism, specific receptor ligands). The clinical presentation of endocrine diseases encompasses a broad spectrum of sign and symptoms. Moreover, endocrine tumors show varying degrees of aggressiveness from well differentiated and indolent to highly aggressive cancers, respectively. RATIONALE With the application of new medicinal radio-compounds and increasingly advanced tomographic imaging technology, the utility of Positron Emission Tomography/Computed Tomography (PET/CT) in the field of endocrine diseases is expanding. AIM This review aims to analyze and summarize the primary indications of PET/CT, providing a practical approach for clinicians. A comprehensive literature search on PubMed was conducted to provide an updated overview of the available evidence regarding the use of PET/CT in endocrinology. Within this review, we will discuss the applications of PET/CT, compare different radiopharmaceuticals and highlight the uptake mechanism, excluding neuroendocrine carcinomas from discussion. CONCLUSIONS PET/CT is a valuable tool in diagnosing and managing endocrine disorders due to its capacity to furnish both functional and anatomical information, facilitate early lesion detection, guide treatment decisions, and monitor treatment response. Its non-invasive nature and precision make it an integral component of modern endocrine healthcare. This review aims to provide physicians with a clear perspective on the role of PET/CT imaging, discussing its emerging opportunities and appropriateness of use in endocrinological diseases.
Collapse
Affiliation(s)
- L Calderoni
- Nuclear Medicine Division, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico di S. Orsola, Via Albertoni 15, Bologna, Italy.
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126, Bologna, Italy.
| | - L Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Clinic for Nuclear Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - S Fanti
- Nuclear Medicine Division, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico di S. Orsola, Via Albertoni 15, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126, Bologna, Italy
| |
Collapse
|
5
|
Shah S, Lucke-Wold B. Image-Guided Mesenchymal Stem Cell Sodium Iodide Symporter (NIS) Radionuclide Therapy for Glioblastoma. Cancers (Basel) 2024; 16:2892. [PMID: 39199662 PMCID: PMC11352884 DOI: 10.3390/cancers16162892] [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: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive, invasive, and growth factor-independent grade IV glioma. Survival following the diagnosis is generally poor, with a median survival of approximately 15 months, and it is considered the most aggressive and lethal central nervous system tumor. Conventional treatments based on surgery, chemotherapy, and radiation therapy only delay progression, and death is inevitable. Malignant glioma cells are resistant to traditional therapies, potentially due to a subpopulation of glioma stem cells that are invasive and capable of rapid regrowth. METHODS This is a literature review. The systematic retrieval of information was performed on PubMed, Embase, and Google Scholar. Specified keywords were used in PubMed and the articles retrieved were published in peer-reviewed scientific journals and were associated with brain GBM cancer and the sodium iodide symporter (NIS). Additionally, the words 'radionuclide therapy OR mesenchyma, OR radioiodine OR iodine-131 OR molecular imaging OR gene therapy OR translational imaging OR targeted OR theranostic OR symporter OR virus OR solid tumor OR combined therapy OR pituitary OR plasmid AND glioblastoma OR GBM OR GB OR glioma' were also used in the appropriate literature databases of PubMed and Google Scholar. A total of 68,244 articles were found in this search on Mesenchymal Stem Cell Sodium Iodide Symporter and GBM. These articles were found till 2024. To study recent advances, a filter was added to include articles only from 2014 to 2024, duplicates were removed, and articles not related to the title were excluded. These came out to be 78 articles. From these, nine were not retrieved and only seven were selected after the removal of keyword mismatched articles. Appropriate studies were isolated, and important information from each of them was understood and entered into a database from which the information was used in this article. RESULTS As a result of their natural capacity to identify malignancies, MSCs are employed as tumor therapy vehicles. Because MSCs may be transplanted using several methods, they have been proposed as the ideal vehicles for NIS gene transfer. MSCs have been used as a delivery vector for anticancer drugs in many tumor models due to their capacity to move precisely to malignancies. Also, by directly injecting radiolabeled MSCs into malignant tumors, a therapeutic dosage of beta radiation may be deposited, with the added benefit that the tumor would only localize and not spread to the surrounding healthy tissues. CONCLUSION The non-invasive imaging-based detection of glioma stem cells presents an alternate means to monitor the tumor and diagnose and evaluate recurrence. The sodium iodide symporter gene is a specific gene in a variety of human thyroid diseases that functions to move iodine into the cell. In recent years, an increasing number of studies related to the sodium iodide symporter gene have been reported in a variety of tumors and as therapeutic vectors for imaging and therapy. Gene therapy and nuclear medicine therapy for GBM provide a new direction. In all the preclinical studies reviewed, image-guided cell therapy led to greater survival benefits and, therefore, has the potential to be translated into techniques in glioblastoma treatment trials.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA;
| | | |
Collapse
|
6
|
Petranović Ovčariček P, Calderoni L, Campenni A, Fanti S, Giovanella L. Molecular imaging of thyroid and parathyroid diseases. Expert Rev Endocrinol Metab 2024; 19:317-333. [PMID: 38899737 DOI: 10.1080/17446651.2024.2365776] [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: 03/24/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Molecular imaging of thyroid and parathyroid diseases has changed in recent years due to the introduction of new radiopharmaceuticals and new imaging techniques. Accordingly, we provided an clinicians-oriented overview of such techniques and their indications. AREAS COVERED A review of the literature was performed in the PubMed, Web of Science, and Scopus without time or language restrictions through the use of one or more fitting search criteria and terms as well as through screening of references in relevant selected papers. Literature up to and including December 2023 was included. Screening of titles/abstracts and removal of duplicates was performed and the full texts of the remaining potentially relevant articles were retrieved and reviewed. EXPERT OPINION Thyroid and parathyroid scintigraphy remains integral in patients with thyrotoxicosis, thyroid nodules, differentiated thyroid cancer and, respectively, hyperparathyroidism. In the last years positron-emission tomography with different tracers emerged as a more accurate alternative in evaluating indeterminate thyroid nodules [18F-fluorodeoxyglucose (FDG)], differentiated thyroid cancer [124I-iodide, 18F-tetrafluoroborate, 18F-FDG] and hyperparathyroidism [18F-fluorocholine]. Other PET tracers are useful in evaluating relapsing/advanced forms of medullary thyroid cancer (18F-FDOPA) and selecting patients with advanced follicular and medullary thyroid cancers for theranostic treatments (68Ga/177Ga-somatostatin analogues).
Collapse
Affiliation(s)
- Petra Petranović Ovčariček
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Letizia Calderoni
- Nuclear Medicine Division, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico S. Orsola, Bologna, Italy
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alfredo Campenni
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Unit of Nuclear Medicine, University of Messina, Messina, Italy
| | - Stefano Fanti
- Nuclear Medicine Division, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico S. Orsola, Bologna, Italy
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Luca Giovanella
- Department of Nuclear Medicine, Gruppo Ospedaliero Moncucco, Lugano, Switzerland
- Clinic for Nuclear Medicine, University Hospital of Zürich, Zürich, Switzerland
| |
Collapse
|
7
|
Mulugeta PG, Chi AW, Anderson TM. Molecular Imaging and Therapy of Differentiated Thyroid Carcinoma in Adults. Cancer J 2024; 30:194-201. [PMID: 38753754 DOI: 10.1097/ppo.0000000000000713] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Differentiated thyroid carcinoma (DTC) has been increasing in incidence in the United States over the last several decades, although mortality rates have remained low. Radioactive iodine therapy (RAI-T) has been a mainstay of treatment for DTC since the 1940s. Imaging of DTC before and after RAI-T primarily focuses on molecular imaging of the sodium iodide symporter. The expanding understanding of the molecular profile of DTC has increased available treatment options. Incorporation of risk stratification to treatment approaches has led to deintensification of both surgical and nonsurgical treatments, leading to decreased morbidity without compromising disease control.
Collapse
Affiliation(s)
- Philipose Getachew Mulugeta
- From the Associate Professor of Clinical Radiology, Clinical Director, Division of Nuclear Medicine Imaging and Therapy, Hospital of the University of Pennsylvania, Department of Radiology, 3400 Spruce Street, 1 Silverstein
| | - Anthony W Chi
- Staff Pathologist, Subchief for Molecular Pathology, Head & Neck Pathology and Hematology, Mid-Atlantic Permanente Medical Group, Regional Laboratory, 611 Executive Blvd, Rockville, MD 20852; and
| | - Thomas Michael Anderson
- Assistant Professor, Director of Therapeutic Nuclear Medicine, Department of Radiology, UNM School of Medicine, MSC10 5530, 1 University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
8
|
Chandekar KR, Satapathy S, Bal C. Positron Emission Tomography/Computed Tomography in Thyroid Cancer: An Updated Review. PET Clin 2024; 19:131-145. [PMID: 38212213 DOI: 10.1016/j.cpet.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
PET/computed tomography (CT) is a valuable hybrid imaging modality for the evaluation of thyroid cancer, potentially impacting management decisions. 18F-fluorodeoxyglucose (FDG) PET/CT has proven utility for recurrence evaluation in differentiated thyroid cancer (DTC) patients having thyroglobulin elevation with negative iodine scintigraphy. Aggressive histologic subtypes such as anaplastic thyroid cancer shower higher FDG uptake. 18F-FDOPA is the preferred PET tracer for medullary thyroid cancer. Fibroblast activation protein inhibitor and arginylglycylaspartic acid -based radiotracers have emerged as promising PET agents for radioiodine refractory DTC patients with the potential for theranostic application.
Collapse
|
9
|
Bowden GD, Stotz S, Dunkel G, Haas S, Kimmerle E, Schaller M, Weigelin B, Herfert K, Pichler BJ, Maurer A. [ 18F] pFBC, a Covalent CLIP-Tag Radiotracer for Detection of Viral Reporter Gene Transfer in the Murine Brain. Bioconjug Chem 2024; 35:254-264. [PMID: 38308817 DOI: 10.1021/acs.bioconjchem.3c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2024]
Abstract
Preclinical models of neurological diseases and gene therapy are essential for neurobiological research. However, the evaluation of such models lacks reliable reporter systems for use with noninvasive imaging methods. Here, we report the development of a reporter system based on the CLIP-tag enzyme and [18F]pFBC, an 18F-labeled covalent CLIP-tag-ligand synthesized via a DoE-optimized and fully automated process. We demonstrated its specificity using a subcutaneous xenograft model and a model of viral vector-mediated brain gene transfer by engineering HEK293 cells and striatal neurons to express membrane-tethered CLIP-tag protein. After in vitro characterization of the reporter, mice carrying either CLIP-tag expressing or control subcutaneous xenografts underwent dynamic [18F]pFBC PET imaging. The CLIP-tag expressing xenografts showed a significantly higher uptake than control xenografts (tumor-to-muscle ratio 5.0 vs 1.7, p = 0.0379). In vivo, metabolite analysis by radio-HPLC from plasma and brain homogenates showed only one radio-metabolite in plasma and none in the brain. In addition, [18F]pFBC showed fast uptake and rapid clearance from the brain in animals injected with adeno-associated virus (AAV)-CLIP in the right striatum but no right-to-left (R-L) uptake difference in the striata in the acquired PET data. In contrast, autoradiography showed a clear accumulation of radioactivity in the AAV-CLIP-injected right striatum compared to the sham-injected left striatum control. CLIP-tag expression and brain integrity were verified by immunofluorescence and light sheet microscopy. In conclusion, we established a novel reporter gene system for PET imaging of gene expression in the brain and periphery and demonstrated its potential for a wide range of applications, particularly for neurobiological research and gene therapy with viral vectors.
Collapse
Affiliation(s)
- Gregory D Bowden
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, 72076 Tuebingen, Germany
| | - Sophie Stotz
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, 72076 Tuebingen, Germany
| | - Gina Dunkel
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, 72076 Tuebingen, Germany
| | - Sabrina Haas
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Elena Kimmerle
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Martin Schaller
- Department of Dermatology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Bettina Weigelin
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, 72076 Tuebingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, 72076 Tuebingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, 72076 Tuebingen, Germany
| |
Collapse
|
10
|
Ventura D, Dittmann M, Büther F, Schäfers M, Rahbar K, Hescheler D, Claesener M, Schindler P, Riemann B, Seifert R, Roll W. Diagnostic Performance of [ 18F]TFB PET/CT Compared with Therapeutic Activity [ 131I]Iodine SPECT/CT and [ 18F]FDG PET/CT in Recurrent Differentiated Thyroid Carcinoma. J Nucl Med 2024; 65:192-198. [PMID: 38164565 PMCID: PMC10858375 DOI: 10.2967/jnumed.123.266513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/01/2023] [Indexed: 01/03/2024] Open
Abstract
[18F]tetrafluoroborate ([18F]TFB) is an emerging PET tracer with excellent properties for human sodium iodide symporter (NIS)-based imaging in patients with differentiated thyroid cancer (DTC). The aim of this study was to compare [18F]TFB PET with high-activity posttherapeutic [131I]iodine whole-body scintigraphy and SPECT/CT in recurrent DTC and with [18F]FDG PET/CT in suspected dedifferentiation. Methods: Twenty-six patients treated with high-activity radioactive [131I]iodine therapy (range, 5.00-10.23 GBq) between May 2020 and November 2022 were retrospectively included. Thyroid-stimulating hormone was stimulated by 2 injections of recombinant thyroid-stimulating hormone (0.9 mg) 48 and 24 h before therapy. Before treatment, all patients underwent [18F]TFB PET/CT 40 min after injection of a median of 321 MBq of [18F]TFB. To study tracer kinetics in DTC lesions, 23 patients received an additional scan at 90 min. [131I]iodine therapeutic whole-body scintigraphy and SPECT/CT were performed at a median of 3.8 d after treatment. Twenty-five patients underwent additional [18F]FDG PET. Two experienced nuclear medicine physicians evaluated all imaging modalities in consensus. Results: A total of 62 suspected lesions were identified; of these, 30 lesions were [131I]iodine positive, 32 lesions were [18F]TFB positive, and 52 were [18F]FDG positive. Three of the 30 [131I]iodine-positive lesions were retrospectively rated as false-positive iodide uptake. Tumor-to-background ratio measurements at the 40- and 90-min time points were closely correlated (e.g., for the tumor-to-background ratio for muscle, the Pearson correlation coefficient was 0.91; P < 0.001; n = 49). We found a significant negative correlation between [18F]TFB uptake and [18F]FDG uptake as a potential marker for dedifferentiation (Pearson correlation coefficient, -0.26; P = 0.041; n = 62). Conclusion: Pretherapeutic [18F]TFB PET/CT may help to predict the positivity of recurrent DTC lesions on [131I]iodine scans. Therefore, it may help in the selection of patients for [131I]iodine therapy. Future prospective trials for iodine therapy guidance are warranted. Lesion [18F]TFB uptake seems to be inversely correlated with [18F]FDG uptake and therefore might serve as a dedifferentiation marker in DTC.
Collapse
Affiliation(s)
- David Ventura
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany;
- West German Cancer Centre, Münster, Germany
| | - Matthias Dittmann
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- Department of Nuclear Medicine, St. Marien Hospital Lünen, Lünen, Germany
| | - Florian Büther
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- West German Cancer Centre, Münster, Germany
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- West German Cancer Centre, Münster, Germany
| | - Daniel Hescheler
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- West German Cancer Centre, Münster, Germany
| | - Michael Claesener
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Philipp Schindler
- West German Cancer Centre, Münster, Germany
- Clinic for Radiology, University and University Hospital Münster, Münster, Germany
| | - Burkhard Riemann
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- West German Cancer Centre, Münster, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- West German Cancer Centre, Münster, Germany
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; and
- West German Cancer Centre, Essen, Germany
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
- West German Cancer Centre, Münster, Germany
| |
Collapse
|
11
|
Garcia N, Ulin M, Yang Q, Ali M, Bosland MC, Zeng W, Chen L, Al-Hendy A. Survivin-Sodium Iodide Symporter Reporter as a Non-Invasive Diagnostic Marker to Differentiate Uterine Leiomyosarcoma from Leiomyoma. Cells 2023; 12:2830. [PMID: 38132150 PMCID: PMC10741838 DOI: 10.3390/cells12242830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Leiomyosarcoma (LMS) has been challenging to diagnose because of limitations in clinical and radiographic predictors, as well as the lack of reliable serum or urinary biomarkers. Most uterine masses consist of benign leiomyoma (LM). However, it is currently a significant challenge in gynecology practice to differentiate LMS from LM. This inability poses grave consequences for patients, leading to a high number of unnecessary hysterectomies, infertility, and other major morbidities and possible mortalities. This study aimed to evaluate the use of Survivin-Sodium iodide symporter (Ad-Sur-NIS) as a reporter gene biomarker to differentiate malignant LMS from benign LM by using an F18-NaBF4 PET/CT scan. The PET/CT scan images showed a significantly increased radiotracer uptake and a decreased radiotracer decay attributable to the higher abundance of Ad-Sur-NIS in the LMS tumors compared to LM (p < 0.05). An excellent safety profile was observed, with no pathological or metabolic differences detected in Ad-Sur-NIS-treated animal versus the vehicle control. Ad-Sur-NIS as a PET scan reporter is a promising imaging biomarker that can differentiate uterine LMS from LM using F18-NaBF4 as a radiotracer. As a new diagnostic method, the F18 NaBF4 PET/CT scan can provide a much-needed tool in clinical practices to effectively triage women with suspicious uterine masses and avoid unnecessary invasive interventions.
Collapse
Affiliation(s)
- Natalia Garcia
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Greehey Children’s Cancer Research Institute, The University of Texas Health Science Center, San Antonio, TX 77030, USA
| | - Mara Ulin
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Chicago, IL 11537, USA
| | - Qiwei Yang
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| | - Mohamed Ali
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Maarten C. Bosland
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Weiqiao Zeng
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
| | - Liaohai Chen
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
| | - Ayman Al-Hendy
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
12
|
Léost F, Barbet J, Beyler M, Chérel M, Delpon G, Garcion E, Lacerda S, Lepareur N, Rbah-Vidal L, Vaugier L, Visvikis D. ["New Modalities in Cancer Imaging and Therapy" XVth edition of the workshop organized by the network "Tumor Targeting, Imaging, Radiotherapies" of the Cancéropôle Grand-Ouest, 5-8 October 2022, France]. Bull Cancer 2023; 110:1322-1331. [PMID: 37880044 DOI: 10.1016/j.bulcan.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/16/2023] [Accepted: 08/13/2023] [Indexed: 10/27/2023]
Abstract
The fifteenth edition of the international workshop organized by the "Tumour Targeting and Radiotherapies network" of the Cancéropôle Grand-Ouest focused on the latest advances in internal and external radiotherapy from different disciplinary angles: chemistry, biology, physics, and medicine. The workshop covered several deliberately diverse topics: the role of artificial intelligence, new tools for imaging and external radiotherapy, theranostic aspects, molecules and contrast agents, vectors for innovative combined therapies, and the use of alpha particles in therapy.
Collapse
Affiliation(s)
- Françoise Léost
- Cancéropôle Grand-Ouest, IRS-UN, 8, quai Moncousu, 44007 Nantes cedex 1, France.
| | | | - Maryline Beyler
- Université de Brest, UMR CNRS-UBO 6521 CEMCA, 6, avenue V.-Le-Gorgeu, 29200 Brest, France
| | - Michel Chérel
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI(2)NA, Nantes, France
| | - Grégory Delpon
- Institut de cancérologie de l'Ouest, département de physique médicale, boulevard Jacques-Monod, 44800 Saint-Herblain, France; Laboratoire SUBATECH, UMR 6457 CNRS-IN2P3, IMT Atlantique, 4, rue Alfred-Kastler, 44307 Nantes cedex 3, France
| | - Emmanuel Garcion
- Université d'Angers, Inserm, CNRS, Nantes Université, CRCI(2)NA, Angers, France
| | - Sara Lacerda
- Université d'Orléans, centre de biophysique moléculaire, CNRS UPR 4301, rue Charles-Sadron, 45071 Orléans cedex 2, France
| | - Nicolas Lepareur
- Université de Rennes, Inrae, Inserm, CLCC Eugène-Marquis, institut nutrition, métabolismes et cancer (NUMECAN), UMR 1317, Rennes, France
| | - Latifa Rbah-Vidal
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI(2)NA, Nantes, France
| | - Loïg Vaugier
- Institut de cancérologie de l'Ouest, département de physique médicale, boulevard Jacques-Monod, 44800 Saint-Herblain, France; Laboratoire SUBATECH, UMR 6457 CNRS-IN2P3, IMT Atlantique, 4, rue Alfred-Kastler, 44307 Nantes cedex 3, France
| | - Dimitris Visvikis
- Inserm, LaTIM, UMR 1101, IBSAM, UBO, UBL, 22, rue Camille-Desmoulins, 29238 Brest, France
| |
Collapse
|
13
|
Saito Y, Nose N, Iida T, Akazawa K, Kanno T, Fujimoto Y, Sasaki T, Akehi M, Higuchi T, Akagi S, Yoshida M, Miyoshi T, Ito H, Nakamura K. In vivo tracking transplanted cardiomyocytes derived from human induced pluripotent stem cells using nuclear medicine imaging. Front Cardiovasc Med 2023; 10:1261330. [PMID: 37745108 PMCID: PMC10512708 DOI: 10.3389/fcvm.2023.1261330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Transplantation of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is a promising treatment for heart failure. Information on long-term cell engraftment after transplantation is clinically important. However, clinically applicable evaluation methods have not yet been established. Methods In this study, to noninvasively assess transplanted cell engraftment, human SLC5A5, which encodes a sodium/iodide symporter (NIS) that transports radioactive tracers such as 125I, 18F-tetrafluoroborate (TFB), and 99mTc-pertechnetate (99mTcO4-), was transduced into human induced pluripotent stem cells (iPSCs), and nuclear medicine imaging was used to track engrafted human iPSC-CMs. Results To evaluate the pluripotency of NIS-expressing human iPSCs, they were subcutaneously transplanted into immunodeficient rats. Teratomas were detected by 99mTcO4- single photon emission computed tomography (SPECT/CT) imaging. NIS expression and the uptake ability of 125I were maintained in purified human iPSC-CMs. NIS-expressing human iPSC-CMs transplanted into immunodeficient rats could be detected over time using 99mTcO4- SPECT/CT imaging. Unexpectedly, NIS expression affected cell proliferation of human iPSCs and iPSC-derived cells. Discussion Such functionally designed iPSC-CMs have potential clinical applications as a noninvasive method of grafted cell evaluation, but further studies are needed to determine the effects of NIS transduction on cellular characteristics and functions.
Collapse
Affiliation(s)
- Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama, Japan
| | - Naoko Nose
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshihiro Iida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kaoru Akazawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takayuki Kanno
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuki Fujimoto
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takanori Sasaki
- Okayama Medical Innovation Center, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaru Akehi
- Okayama Medical Innovation Center, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takahiro Higuchi
- Molecular Imaging Project of RECTOR Program, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masashi Yoshida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Ito
- Department of General Internal Medicine 3, Kawasaki Medical School, Okayama, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
14
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
15
|
Eilsberger F, Kreissl MC, Luster M, Pfestroff A. [Therapy concepts for thyroid carcinoma]. Laryngorhinootologie 2023. [PMID: 37011888 DOI: 10.1055/a-1861-7379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Theranostics via the sodium iodide symporter (NIS) offer a unique option in differentiated thyroid carcinoma. The diagnostic and therapeutic nuclides have similar uptake and kinetics, making the NIS the most important theranostic target in this disease. Radioiodine refractory thyroid carcinomas (RRTC) are characterised by reduced/absent NIS expression, thus eliminating this structure as a theranostic target. Also due to limited therapeutic options, there are approaches to generate new theranostic targets in RRTC, via the expression of somatostatin receptors (SSTR) or the prostate-specific membrane antigen (PSMA), but the current evidence does not yet allow a final evaluation of the prospects of success.
Collapse
Affiliation(s)
| | - Michael C Kreissl
- Abteilung für Nuklearmedizin, Universitatsklinikum Magdeburg, Magdeburg, Germany
| | - Markus Luster
- Nuclearmedicine, University of Marburg, Marburg, Germany
| | - Andreas Pfestroff
- Klinik für Nuklearmedizin, Universitätsklinikum Marburg, Marburg, Germany
| |
Collapse
|
16
|
Role of PET/Computed Tomography in Elderly Thyroid Cancer: Tumor Biology and Clinical Management. PET Clin 2023; 18:81-101. [PMID: 36718717 DOI: 10.1016/j.cpet.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PET/computed tomography (CT) studies can be potentially useful in elderly thyroid carcinoma patients for exploring the disease biology, especially in metastatic setting and thereby directing appropriate therapeutic management on case-to-case basis, adopting nuclear theranostics, and disease prognostication. With the availability of multiple PET radiopharmaceuticals, it would be worthwhile to evolve and optimally use FDG and the other non-fluorodeoxyglucose and investigational PET/CT tracers as per the clinical situation and need and thereby define their utilities in a given case scenario. In this regard, (I) differentiated thyroid carcinoma (DTC) including radioiodine refractory disease, poorly differentiated thyroid cancer (PDTC) and TENIS, (II) medullary thyroid carcinoma (MTC), (III) anaplastic carcinoma and (IV) Primary thyroid lymphoma (PTL) should be viewed and dealt separately.
Collapse
|
17
|
Soloviev D, Dzien P, Mackintosh A, Malviya G, Brown G, Lewis D. High molar activity [ 18F]tetrafluoroborate synthesis for sodium iodide symporter imaging by PET. EJNMMI Radiopharm Chem 2022; 7:32. [PMID: 36512196 PMCID: PMC9747990 DOI: 10.1186/s41181-022-00185-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sodium iodide symporter (NIS) imaging by positron emission tomography (PET) is gaining traction in nuclear medicine, with an increasing number of human studies being published using fluorine-18 radiolabelled tetrafluoroborate ([18F]TFB). Clinical success of any radiotracer relies heavily on its accessibility, which in turn depends on the availability of robust radiolabelling procedures providing a radiotracer in large quantities and of high radiopharmaceutical quality. RESULTS Here we publish an improved radiolabelling method and quality control procedures for high molar activity [18F]TFB. The use of ammonium hydroxide for [18F]fluoride elution, commercially available boron trifluoride-methanol complex dissolved in acetonitrile as precursor and removal of unreacted [18F]fluoride on Florisil solid-phase extraction cartridges resulted in the reliable production of [18F]TFB on SYNTHRA and TRACERLAB FXFN automated synthesizers with radiochemical yields in excess of 30%, radiochemical purities in excess of 98% and molar activities in the range of 34-217 GBq/µmol at the end of synthesis. PET scanning of a mouse lung tumour model carrying a NIS reporter gene rendered images of high quality and improved sensitivity. CONCLUSIONS A novel automated radiosynthesis procedure for [18F]tetrafluoroborate has been developed that provides the radiotracer with high molar activity, suitable for preclinical imaging of NIS reporter gene.
Collapse
Affiliation(s)
- Dmitry Soloviev
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
| | - Piotr Dzien
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Agata Mackintosh
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Gaurav Malviya
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Gavin Brown
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David Lewis
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G611QH, UK
| |
Collapse
|
18
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
19
|
Giovanella L, Avram AM, Ovčariček PP, Clerc J. Thyroid functional and molecular imaging. Presse Med 2022; 51:104116. [PMID: 35124101 DOI: 10.1016/j.lpm.2022.104116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/02/2021] [Accepted: 01/28/2022] [Indexed: 12/16/2022] Open
Abstract
Radioiodine uptake (RAIU) test with iodine-123 (Na[123I]I) or iodine-131 (Na[131I]I) enables accurate evaluation and quantification of iodine uptake and kinetics within thyroid cells. Thyroid Scintigraphy (TS) employing Na[123I]I or 99mTc-pertechnetate (Na[99mTc]TcO4) provides information regarding the function and topographical distribution of thyroid cells activity, including detection and localization of ectopic thyroid tissue. Destructive thyrotoxicosis is characterized by low RAIU with scintigraphically reduced radiotracer activity in the thyroid tissue, while productive thyrotoxicosis (i.e. hyperthyroidism "stricto sensu") is characterized by high RAIU with scintigraphically diffuse (i.e. Graves' Disease, GD and diffuse thyroid autonomy) or focal (i.e. autonomously functioning thyroid nodules, AFTN) overactivity. Accordingly, RAIU and/or TS are widely used to differentiate different causes of thyrotoxicosis. In addition, several radiopharmaceuticals are also available to help differentiate benign from malignant thyroid nodules and inform clinical decision-making: scintigraphic identification of AFTNs obviate fine-needle aspiration (FNA) biopsy, and [99mTc]Tc-hexakis-(2‑methoxy-2-isobutyl isonitrile ([99mTc]Tc-MIBI) and/or 18F-fluoro-d-glucose ([18F]FDG) may complement the work-up of cytologically indeterminate "cold" nodules for reducing the need for diagnostic lobectomies/thyroidectomies. Finally, RAIU studies are also useful for calculating the administered therapeutic activity of Na[131I]I to treat hyperthyroidism and euthyroid multinodular goiter. All considered, thyroid molecular imaging allows functional characterization of different thyroid diseases, even before clinical symptoms become manifest, and remains integral to the management of such conditions. Our present paper summarizes basic concepts, clinical applications, and potential developments of thyroid molecular imaging in patients affected by thyrotoxicosis and thyroid nodules.
Collapse
Affiliation(s)
- Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Clinic for Nuclear Medicine, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Anca M Avram
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Division of Endocrinology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Petra Petranović Ovčariček
- Department of Oncology and Nuclear Medicine, University Hospital Center "Sestre milosrdnice", Zagreb, Croatia
| | - Jerome Clerc
- Department of Nuclear Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, DMU Imagina, University of Paris France
| |
Collapse
|
20
|
Schlumberger M, Garcia C, Hadoux J, Klain M, Lamartina L. Functional imaging in thyroid cancer patients with metastases and therapeutic implications. Presse Med 2022; 51:104113. [PMID: 35131318 DOI: 10.1016/j.lpm.2022.104113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/26/2021] [Accepted: 01/28/2022] [Indexed: 12/16/2022] Open
Abstract
Functional imaging plays a central role in the management of thyroid cancer patients. In patients with a differentiated thyroid cancer (DTC), radioactive iodine (RAI) is used mostly with a therapeutic intent, either post-operatively or as the first line systemic treatment in patients with known structural disease. A whole body scan is performed a few days after the RAI administration, and this procedure is very sensitive to detect all tumor foci with RAI uptake. PET/CT with 18F-FDG complements the use of RAI at the initial evaluation of patients with high-risk DTC, during follow-up in those with rising serum thyroglobulin levels over time, for the work-up of patients with documented structural disease and for assessing the efficacy of focal or systemic treatment modalities. 18F-FDG uptake is a prognostic indicator in all these clinical conditions. A dosimetric approach with 124I PET/CT showed encouraging results. Several functional imaging modalities are currently available for medullary thyroid carcinoma (MTC) patients. 18F-FDG-PET/CT may be sensitive in MTC patients with high FDG uptake that signals aggressive disease. 18F-DOPA is the most sensitive imaging technique to visualize small tumor foci, and is also highly specific in patients with a known MTC, but should be complemented by a CT scan of the chest and by a MRI of the liver to detect small metastases.
Collapse
Affiliation(s)
- Martin Schlumberger
- Gustave Roussy, Service de Médecine Nucléaire et de Cancérologie Endocrinienne, 114 rue Edouard Vaillant, Villejuif 94800, France.
| | - Camilo Garcia
- Gustave Roussy, Service de Médecine Nucléaire et de Cancérologie Endocrinienne, 114 rue Edouard Vaillant, Villejuif 94800, France
| | - Julien Hadoux
- Gustave Roussy, Service de Médecine Nucléaire et de Cancérologie Endocrinienne, 114 rue Edouard Vaillant, Villejuif 94800, France
| | - Michele Klain
- Gustave Roussy, Service de Médecine Nucléaire et de Cancérologie Endocrinienne, 114 rue Edouard Vaillant, Villejuif 94800, France
| | - Livia Lamartina
- Gustave Roussy, Service de Médecine Nucléaire et de Cancérologie Endocrinienne, 114 rue Edouard Vaillant, Villejuif 94800, France
| |
Collapse
|
21
|
Eilsberger F, Kreissl MC, Luster M, Pfestroff A. [Therapy concepts for thyroid carcinoma]. Nuklearmedizin 2022; 61:223-230. [PMID: 34644802 DOI: 10.1055/a-1650-9762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Theranostics via the sodium iodide symporter (NIS) offer a unique option in differentiated thyroid carcinoma. The diagnostic and therapeutic nuclides have similar uptake and kinetics, making the NIS the most important theranostic target in this disease. Radioiodine refractory thyroid carcinomas (RRTC) are characterised by reduced/absent NIS expression, thus eliminating this structure as a theranostic target. Also due to limited therapeutic options, there are approaches to generate new theranostic targets in RRTC, via the expression of somatostatin receptors (SSTR) or the prostate-specific membrane antigen (PSMA), but the current evidence does not yet allow a final evaluation of the prospects of success.
Collapse
Affiliation(s)
| | - Michael C Kreissl
- Abteilung für Nuklearmedizin, Universitatsklinikum Magdeburg, Magdeburg, Germany
| | - Markus Luster
- Klinik für Nuklearmedizin, Universitätsklinikum Marburg, Marburg, Germany
| | - Andreas Pfestroff
- Klinik für Nuklearmedizin, Universitätsklinikum Marburg, Marburg, Germany
| |
Collapse
|
22
|
Clinical activity of single-dose systemic oncolytic VSV virotherapy in patients with relapsed refractory T-cell lymphoma. Blood Adv 2022; 6:3268-3279. [PMID: 35175355 PMCID: PMC9198941 DOI: 10.1182/bloodadvances.2021006631] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
Clinical success with intravenous (IV) oncolytic virotherapy (OV) has to-date been anecdotal. We conducted a phase 1 clinical trial of systemic OV and investigated the mechanisms of action in responding patients. A single IV dose of vesicular stomatitis virus (VSV) interferon-β (IFN-β) with sodium iodide symporter (NIS) was administered to patients with relapsed/refractory hematologic malignancies to determine safety and efficacy across 4 dose levels (DLs). Correlative studies were undertaken to evaluate viremia, virus shedding, virus replication, and immune responses. Fifteen patients received VSV-IFNβ-NIS. Three patients were treated at DL1 through DL3 (0.05, 0.17, and 0.5 × 1011 TCID50), and 6 were treated at DL4 (1.7 × 1011 TCID50) with no dose-limiting toxicities. Three of 7 patients with T-cell lymphoma (TCL) had responses: a 3-month partial response (PR) at DL2, a 6-month PR, and a complete response (CR) ongoing at 20 months at DL4. Viremia peaked at the end of infusion, g was detected. Plasma IFN-β, a biomarker of VSV-IFNβ-NIS replication, peaked between 4 hours and 48 hours after infusion. The patient with CR had robust viral replication with increased plasma cell-free DNA, high peak IFN-β of 18 213 pg/mL, a strong anti-VSV neutralizing antibody response, and increased numbers of tumor reactive T-cells. VSV-IFNβ-NIS as a single agent was effective in patients with TCL, resulting in durable disease remissions in heavily pretreated patients. Correlative analyses suggest that responses may be due to a combination of direct oncolytic tumor destruction and immune-mediated tumor control. This trial is registered at www.clinicaltrials.gov as #NCT03017820.
Collapse
|
23
|
Kitzberger C, Spellerberg R, Morath V, Schwenk N, Schmohl KA, Schug C, Urnauer S, Tutter M, Eiber M, Schilling F, Weber WA, Ziegler S, Bartenstein P, Wagner E, Nelson PJ, Spitzweg C. The sodium iodide symporter (NIS) as theranostic gene: its emerging role in new imaging modalities and non-viral gene therapy. EJNMMI Res 2022; 12:25. [PMID: 35503582 PMCID: PMC9065223 DOI: 10.1186/s13550-022-00888-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/11/2022] [Indexed: 01/14/2023] Open
Abstract
Cloning of the sodium iodide symporter (NIS) in 1996 has provided an opportunity to use NIS as a powerful theranostic transgene. Novel gene therapy strategies rely on image-guided selective NIS gene transfer in non-thyroidal tumors followed by application of therapeutic radionuclides. This review highlights the remarkable progress during the last two decades in the development of the NIS gene therapy concept using selective non-viral gene delivery vehicles including synthetic polyplexes and genetically engineered mesenchymal stem cells. In addition, NIS is a sensitive reporter gene and can be monitored by high resolution PET imaging using the radiotracers sodium [124I]iodide ([124I]NaI) or [18F]tetrafluoroborate ([18F]TFB). We performed a small preclinical PET imaging study comparing sodium [124I]iodide and in-house synthesized [18F]TFB in an orthotopic NIS-expressing glioblastoma model. The results demonstrated an improved image quality using [18F]TFB. Building upon these results, we will be able to expand the NIS gene therapy approach using non-viral gene delivery vehicles to target orthotopic tumor models with low volume disease, such as glioblastoma. Trial registration not applicable.
Collapse
Affiliation(s)
- Carolin Kitzberger
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Rebekka Spellerberg
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Volker Morath
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Kathrin A Schmohl
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Christina Schug
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Sarah Urnauer
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Mariella Tutter
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Centre for System-Based Drug Research and Centre for Nanoscience, LMU Munich, Munich, Germany
| | - Peter J Nelson
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany. .,Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
24
|
Personalized Diagnosis in Differentiated Thyroid Cancers by Molecular and Functional Imaging Biomarkers: Present and Future. Diagnostics (Basel) 2022; 12:diagnostics12040944. [PMID: 35453992 PMCID: PMC9030409 DOI: 10.3390/diagnostics12040944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Personalized diagnosis can save unnecessary thyroid surgeries, in cases of indeterminate thyroid nodules, when clinicians tend to aggressively treat all these patients. Personalized diagnosis benefits from a combination of imagery and molecular biomarkers, as well as artificial intelligence algorithms, which are used more and more in our timeline. Functional imaging diagnosis such as SPECT, PET, or fused images (SPECT/CT, PET/CT, PET/MRI), is exploited at maximum in thyroid nodules, with a long history in the past and a bright future with many suitable radiotracers that could properly contribute to diagnosing malignancy in thyroid nodules. In this way, patients will be spared surgery complications, and apparently more expensive diagnostic workouts will financially compensate each patient and also the healthcare system. In this review we will summarize essential available diagnostic tools for malignant and benignant thyroid nodules, beginning with functional imaging, molecular analysis, and combinations of these two and other future strategies, including AI or NIS targeted gene therapy for thyroid carcinoma diagnosis and treatment as well.
Collapse
|
25
|
Shalaby N, Kelly J, Martinez F, Fox M, Qi Q, Thiessen J, Hicks J, Scholl TJ, Ronald JA. A Human-derived Dual MRI/PET Reporter Gene System with High Translational Potential for Cell Tracking. Mol Imaging Biol 2022; 24:341-351. [PMID: 35146614 PMCID: PMC9235057 DOI: 10.1007/s11307-021-01697-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Reporter gene imaging has been extensively used to longitudinally report on whole-body distribution and viability of transplanted engineered cells. Multi-modal cell tracking can provide complementary information on cell fate. Typical multi-modal reporter gene systems often combine clinical and preclinical modalities. A multi-modal reporter gene system for magnetic resonance imaging (MRI) and positron emission tomography (PET), two clinical modalities, would be advantageous by combining the sensitivity of PET with the high-resolution morphology and non-ionizing nature of MRI. PROCEDURES We developed and evaluated a dual MRI/PET reporter gene system composed of two human-derived reporter genes that utilize clinical reporter probes for engineered cell detection. As a proof-of-concept, breast cancer cells were engineered to co-express the human organic anion transporter polypeptide 1B3 (OATP1B3) that uptakes the clinical MRI contrast agent gadolinium ethoxybenzyl-diethylenetriaminepentaacetic acid (Gd-EOB-DTPA), and the human sodium iodide symporter (NIS) which uptakes the PET tracer, [18F] tetrafluoroborate ([18F] TFB). RESULTS T1-weighted MRI results in mice exhibited significantly higher MRI signals in reporter-gene-engineered mammary fat pad tumors versus contralateral naïve tumors (p < 0.05). No differences in contrast enhancement were observed at 5 h after Gd-EOB-DTPA administration using either intravenous or intraperitoneal injection. We also found significantly higher standard uptake values (SUV) in engineered tumors in comparison to the naïve tumors in [18F]TFB PET images (p < 0.001). Intratumoral heterogeneity in signal enhancement was more conspicuous in relatively higher resolution MR images compared to PET images. CONCLUSIONS Our study demonstrates the ability to noninvasively track cells engineered with our human-derived dual MRI/PET reporter system, enabling a more comprehensive evaluation of transplanted cells. Future work is focused on applying this tool to track therapeutic cells, which may one day enable the broader application of cell tracking within the healthcare system.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.
| | - John Kelly
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Francisco Martinez
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Mathew Fox
- Lawson Health Research Institute, London, Canada
- Saint Joseph's Health Care, Toronto, Canada
| | - Qi Qi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Jonathan Thiessen
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Saint Joseph's Health Care, Toronto, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, Canada
| | - Justin Hicks
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
| | - John A Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
| |
Collapse
|
26
|
Sato N, Choyke PL. Whole-Body Imaging to Assess Cell-Based Immunotherapy: Preclinical Studies with an Update on Clinical Translation. Mol Imaging Biol 2022; 24:235-248. [PMID: 34816284 PMCID: PMC8983636 DOI: 10.1007/s11307-021-01669-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/28/2022]
Abstract
In the past decades, immunotherapies against cancers made impressive progress. Immunotherapy includes a broad range of interventions that can be separated into two major groups: cell-based immunotherapies, such as adoptive T cell therapies and stem cell therapies, and immunomodulatory molecular therapies such as checkpoint inhibitors and cytokine therapies. Genetic engineering techniques that transduce T cells with a cancer-antigen-specific T cell receptor or chimeric antigen receptor have expanded to other cell types, and further modulation of the cells to enhance cancer targeting properties has been explored. Because cell-based immunotherapies rely on cells migrating to target organs or tissues, there is a growing interest in imaging technologies that non-invasively monitor transferred cells in vivo. Here, we review whole-body imaging methods to assess cell-based immunotherapy using a variety of examples. Following a review of preclinically used cell tracking technologies, we consider the status of their clinical translation.
Collapse
Affiliation(s)
- Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10/Rm. B3B406, 10 Center Dr, Bethesda, MD, 20892, USA.
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10/Rm. B3B69F, 10 Center Dr, Bethesda, MD, 20892, USA
| |
Collapse
|
27
|
Giovanella L, Deandreis D, Vrachimis A, Campenni A, Petranovic Ovcaricek P. Molecular Imaging and Theragnostics of Thyroid Cancers. Cancers (Basel) 2022; 14:1272. [PMID: 35267580 PMCID: PMC8909041 DOI: 10.3390/cancers14051272] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging plays an important role in the evaluation and management of different thyroid cancer histotypes. The existing risk stratification models can be refined, by incorporation of tumor-specific molecular markers that have theranostic power, to optimize patient-specific (individualized) treatment decisions. Molecular imaging with varying radioisotopes of iodine (i.e., 131I, 123I, 124I) is an indispensable component of dynamic and theragnostic risk stratification of differentiated carcinoma (DTC) while [18F]F-fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) helps in addressing disease aggressiveness, detects distant metastases, and risk-stratifies patients with radioiodine-refractory DTC, poorly differentiated and anaplastic thyroid cancers. For medullary thyroid cancer (MTC), a neuroendocrine tumor derived from thyroid C-cells, [18F]F-dihydroxyphenylalanine (6-[18F]FDOPA) PET/CT and/or [18F]FDG PET/CT can be used dependent on serum markers levels and kinetics. In addition to radioiodine therapy for DTC, some theragnostic approaches are promising for metastatic MTC as well. Moreover, new redifferentiation strategies are now available to restore uptake in radioiodine-refractory DTC while new theragnostic approaches showed promising preliminary results for advanced and aggressive forms of follicular-cell derived thyroid cancers (i.e., peptide receptor radiotherapy). In order to help clinicians put the role of molecular imaging into perspective, the appropriate role and emerging opportunities for molecular imaging and theragnostics in thyroid cancer are discussed in our present review.
Collapse
Affiliation(s)
- Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Desiree’ Deandreis
- Division of Nuclear Medicine, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy;
| | - Alexis Vrachimis
- Department of Nuclear Medicine, German Oncology Center, University Hospital of the European University, Limassol 4108, Cyprus;
| | - Alfredo Campenni
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98121 Messina, Italy;
| | - Petra Petranovic Ovcaricek
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, 10000 Zagreb, Croatia;
| |
Collapse
|
28
|
Evaluation of [18F]tetrafluoroborate as a Potential PET Imaging Agent in a Sodium Iodide Symporter-Transfected Cell Line A549 and Endogenous NIS-Expressing Cell Lines MKN45 and K1. Mol Imaging 2022; 2022:2679260. [PMID: 35330799 PMCID: PMC8923191 DOI: 10.1155/2022/2679260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
[18F]tetrafluoroborate (TFB) has been introduced as the 18F-labeled PET imaging probe for the human sodium iodide symporter (NIS). Noninvasive NIS imaging using [18F]TFB has received much interest in recent years for evaluating various NIS-expressing tumors. Cancers are a global concern with enormous implications; therefore, improving diagnostic methods for accurate detection of cancer is extremely important. Our aim was to investigate the PET imaging capabilities of [18F]TFB in NIS-transfected lung cell line A549 and endogenous NIS-expressing tumor cells, such as thyroid cancer K1 and gastric cancer MKN45, and broaden its application in the medical field. Western blot and flow cytometry were used to assess the NIS expression level. Radioactivity counts of [18F]TFB, in vitro, in the three tumor cells were substantially higher than those in the KI inhibition group in the uptake experiment. In vivo PET imaging clearly delineated the three tumors based on the specific accumulation of [18F]TFB in a mouse model. Ex vivo biodistribution investigation showed high [18F]TFB absorption in the tumor location, which was consistent with the PET imaging results. These results support the use of NIS-transfected lung cell line A549 and NIS-expressing tumor cells MKN45 and K1, to investigate probing capabilities of [18F]TFB. We also demonstrate, for the first time, the feasibility of [18F]TFB in diagnosing stomach cancer. In conclusion, this study illustrates the promising future of [18F]TFB for tumor diagnosis and NIS reporter imaging.
Collapse
|
29
|
Barca C, Griessinger CM, Faust A, Depke D, Essler M, Windhorst AD, Devoogdt N, Brindle KM, Schäfers M, Zinnhardt B, Jacobs AH. Expanding Theranostic Radiopharmaceuticals for Tumor Diagnosis and Therapy. Pharmaceuticals (Basel) 2021; 15:13. [PMID: 35056071 PMCID: PMC8780589 DOI: 10.3390/ph15010013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
Radioligand theranostics (RT) in oncology use cancer-type specific biomarkers and molecular imaging (MI), including positron emission tomography (PET), single-photon emission computed tomography (SPECT) and planar scintigraphy, for patient diagnosis, therapy, and personalized management. While the definition of theranostics was initially restricted to a single compound allowing visualization and therapy simultaneously, the concept has been widened with the development of theranostic pairs and the combination of nuclear medicine with different types of cancer therapies. Here, we review the clinical applications of different theranostic radiopharmaceuticals in managing different tumor types (differentiated thyroid, neuroendocrine prostate, and breast cancer) that support the combination of innovative oncological therapies such as gene and cell-based therapies with RT.
Collapse
Affiliation(s)
- Cristina Barca
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
| | - Christoph M. Griessinger
- Roche Innovation Center, Early Clinical Development Oncology, Roche Pharmaceutical Research and Early Development, CH-4070 Basel, Switzerland;
| | - Andreas Faust
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Nuclear Medicine, University Hospital Münster, D-48149 Münster, Germany
| | - Dominic Depke
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, D-53127 Bonn, Germany;
| | - Albert D. Windhorst
- Department Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands;
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, B-1090 Brussel, Belgium;
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 ORE, UK;
| | - Michael Schäfers
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Nuclear Medicine, University Hospital Münster, D-48149 Münster, Germany
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Nuclear Medicine, University Hospital Münster, D-48149 Münster, Germany
- Biomarkers and Translational Technologies, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Andreas H. Jacobs
- European Institute for Molecular Imaging, University of Münster, D-48149 Münster, Germany; (A.F.); (D.D.); (M.S.); (B.Z.)
- Department of Geriatrics and Neurology, Johanniter Hospital, D-53113 Bonn, Germany
- Centre of Integrated Oncology, University Hospital Bonn, D-53127 Bonn, Germany
| |
Collapse
|
30
|
Chaurasiya S, Kim SI, O'Leary M, Park AK, Lu J, Kang S, Zhang Z, Yang A, Woo Y, Fong Y, Warner SG. Toward comprehensive imaging of oncolytic viroimmunotherapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:303-310. [PMID: 34786474 PMCID: PMC8569424 DOI: 10.1016/j.omto.2021.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oncolytic viruses infect, replicate in, and kill cancer cells, leaving normal cells unharmed; they also recruit and activate immune cells against tumor cells. While clinical indications for viroimmunotherapy are growing, barriers to widespread treatment remain. Ensuring real-time tracking of viral replication and resulting anti-tumor immune responses will overcome some of these barriers and is thus a top priority. Clinically optimizing trackability of viral replication will promote safe dose increases, guide serial dosing, and enhance treatment effects. However, viral delivery is only half the story. Oncolytic viruses are known to upregulate immune checkpoint expression, thereby priming otherwise immunodeficient tumor immune microenvironments for treatment with checkpoint inhibitors. Novel modalities to track virus-induced changes in tumor microenvironments include non-invasive measurements of immune cell populations and responses to viroimmunotherapy such as (1) in situ use of radiotracers to track checkpoint protein expression or immune cell traffic, and (2) ex vivo labeling of immune cells followed by nuclear medicine imaging. Herein, we review clinical progress toward accurate imaging of oncolytic virus replication, and we further review the current status of functional imaging of immune responses to viroimmunotherapy.
Collapse
Affiliation(s)
- Shyambabu Chaurasiya
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Sang-In Kim
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Michael O'Leary
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Anthony K Park
- Center for Gene Therapy, Department of Hematologic and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jianming Lu
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Seonah Kang
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Zhifang Zhang
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Annie Yang
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| | - Susanne G Warner
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Pavilion 2226, Duarte, CA 91010, USA
| |
Collapse
|
31
|
Advances in Functional Imaging of Differentiated Thyroid Cancer. Cancers (Basel) 2021; 13:cancers13194748. [PMID: 34638232 PMCID: PMC8507556 DOI: 10.3390/cancers13194748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Since the 1940s, radioactive iodine has been used for functional imaging and for treating patients with differentiated thyroid cancer (DTC). During this long-lasting experience, the use of iodine isotopes evolved, especially during the last years due to improved knowledge of thyroid cancer biology and improved performances of imaging tools. The present review summarizes recent advances in the field of functional imaging and theragnostic approach of DTC. Abstract The present review provides a description of recent advances in the field of functional imaging that takes advantage of the functional characteristics of thyroid neoplastic cells (such as radioiodine uptake and FDG uptake) and theragnostic approach of differentiated thyroid cancer (DTC). Physical and biological characteristics of available radiopharmaceuticals and their use with state-of-the-art technologies for diagnosis, treatment, and follow-up of DTC patients are depicted. Radioactive iodine is used mostly with a therapeutic intent, while PET/CT with 18F-FDG emerges as a useful tool in the diagnostic management and complements the use of radioactive iodine. Beyond 18F-FDG PET/CT, other tracers including 124I, 18F-TFB and 68Ga-PSMA, and new methods such as PET/MR, might offer new opportunities in selecting patients with DTC for specific imaging modalities or treatments.
Collapse
|
32
|
Concilio SC, Russell SJ, Peng KW. A brief review of reporter gene imaging in oncolytic virotherapy and gene therapy. Mol Ther Oncolytics 2021; 21:98-109. [PMID: 33981826 PMCID: PMC8065251 DOI: 10.1016/j.omto.2021.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reporter gene imaging (RGI) can accelerate development timelines for gene and viral therapies by facilitating rapid and noninvasive in vivo studies to determine the biodistribution, magnitude, and durability of viral gene expression and/or virus infection. Functional molecular imaging systems used for this purpose can be divided broadly into deep-tissue and optical modalities. Deep-tissue modalities, which can be used in animals of any size as well as in human subjects, encompass single photon emission computed tomography (SPECT), positron emission tomography (PET), and functional/molecular magnetic resonance imaging (f/mMRI). Optical modalities encompass fluorescence, bioluminescence, Cerenkov luminescence, and photoacoustic imaging and are suitable only for small animal imaging. Here we discuss the mechanisms of action and relative merits of currently available reporter gene systems, highlighting the strengths and weaknesses of deep tissue versus optical imaging systems and the hardware/reagents that are used for data capture and processing. In light of recent technological advances, falling costs of imaging instruments, better availability of novel radioactive and optical tracers, and a growing realization that RGI can give invaluable insights across the entire in vivo translational spectrum, the approach is becoming increasingly essential to facilitate the competitive development of new virus- and gene-based drugs.
Collapse
Affiliation(s)
| | | | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
33
|
Jin Y, Liu B, Younis MH, Huang G, Liu J, Cai W, Wei W. Next-Generation Molecular Imaging of Thyroid Cancer. Cancers (Basel) 2021; 13:3188. [PMID: 34202358 PMCID: PMC8268517 DOI: 10.3390/cancers13133188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
An essential aspect of thyroid cancer (TC) management is personalized and precision medicine. Functional imaging of TC with radioiodine and [18F]FDG has been frequently used in disease evaluation for several decades now. Recently, advances in molecular imaging have led to the development of novel tracers based on aptamer, peptide, antibody, nanobody, antibody fragment, and nanoparticle platforms. The emerging targets-including HER2, CD54, SHP2, CD33, and more-are promising targets for clinical translation soon. The significance of these tracers may be realized by outlining the way they support the management of TC. The provided examples focus on where preclinical investigations can be translated. Furthermore, advances in the molecular imaging of TC may inspire the development of novel therapeutic or theranostic tracers. In this review, we summarize TC-targeting probes which include transporter-based and immuno-based imaging moieties. We summarize the most recent evidence in this field and outline how these emerging strategies may potentially optimize clinical practice.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, China
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Beibei Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliatede to Shanghai Jiao Tong University, Shanghai 200233, China;
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| |
Collapse
|
34
|
Zampella E, Klain M, Pace L, Cuocolo A. PET/CT in the management of differentiated thyroid cancer. Diagn Interv Imaging 2021; 102:515-523. [PMID: 33926848 DOI: 10.1016/j.diii.2021.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022]
Abstract
The standard treatment of differentiated thyroid cancer (DTC) consists of surgery followed by iodine-131 (131I) administration. Although the majority of DTC has a very good prognosis, more aggressive histologic subtypes convey a worse prognosis. Follow-up consists of periodically measurements of serum thyroglobulin, thyroglobulin antibodies and neck ultrasound and 123I/131I whole-body scan. However, undifferentiated thyroid tumors have a lower avidity for radioiodine and the ability of DTC to concentrate 131I may be lost in metastatic disease. Positron emission tomography (PET)/computed tomography (CT) has been introduced in the evaluation of patients with thyroid tumors and the 2-[18F]-fluoro-2-deoxyd-glucose (18F-FDG) has been largely validated as marker of cell's metabolism. According to the 2015 American Thyroid Association guidelines, 18F-FDG PET/CT is recommended in the follow-up of high-risk patients with elevated serum thyroglobulin and negative 131I imaging, in the assessment of metastatic patients, for lesion detection and risk stratification and in predicting the response to therapy. It should be considered that well-differentiated iodine avid lesions could not concentrate 18F-FDG, and a reciprocal pattern of iodine and 18F-FDG uptake has been observed. Beyond 18F-FDG, other tracers are available for PET imaging of thyroid tumors, such as Iodine-124 (124I), 18F-tetrafluoroborate and Gallium-68 prostate-specific membrane antigen. Moreover, the recent introduction of PET/MRI, offers now several opportunities in the field of patients with DTC. This review summarizes the evidences on the role of PET/CT in management of patients with DTC, focusing on potential applications and on elucidating some still debating points.
Collapse
Affiliation(s)
- Emilia Zampella
- Department of Advanced Biomedical Sciences, University Federico II, 80131 Naples, Italy.
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University Federico II, 80131 Naples, Italy
| | - Leonardo Pace
- Department of Medicine, Surgery and Dentistry, Università degli Studi di Salerno, 84084 Fisciano, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University Federico II, 80131 Naples, Italy
| |
Collapse
|
35
|
Matsusaka Y, Yamane T, Fukushima K, Seto A, Matsunari I, Kuji I. Can the function of the tubarial glands be evaluated using [ 99mTc]pertechnetate SPECT/CT, [ 18F]FDG PET/CT, and [ 11C]methionine PET/CT? EJNMMI Res 2021; 11:34. [PMID: 33788026 PMCID: PMC8012434 DOI: 10.1186/s13550-021-00779-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The tubarial glands (TGs) are recently reported as newly found salivary gland structures that can be organs at risk predominantly localized in the tori tubarius in the nasopharynx using prostate-specific membrane antigen positron emission tomography/computed tomography (PSMA PET/CT). The aims of this study were to analyze uptake in the TGs compared with that in the other salivary glands and palatine tonsils using [99mTc]pertechnetate SPECT/CT, [18F]FDG PET/CT, and [11C]methionine PET/CT and to confirm whether these three imaging modalities are useful in evaluating the physiological function of the TGs. Twelve and 130 patients, who underwent [99mTc]pertechnetate SPECT/CT and [18F]FDG/[11C]methionine PET/CT, respectively, were retrospectively included. [99mTc]pertechnetate uptake in the tori tubarius was visually assessed and semiquantitatively compared with that in the background, parotid salivary glands (PSGs), submandibular salivary glands (SmSGs), and sublingual salivary glands (SlSGs). Correlations of [18F]FDG and [11C]methionine uptakes in the tori tubarius with those in the other three salivary glands and palatine tonsils were analyzed. RESULTS [99mTc]pertechnetate uptake in the tori tubarius was invisible and was not significantly higher than that in the background. Both [18F]FDG and [11C]methionine uptakes in the tori tubarius were correlated with that in the palatine tonsils (r = 0.56, p < 0.0001; r = 0.48, p < 0.0001, respectively). [18F]FDG uptake in the tori tubarius was not positively correlated with that in the PSGs, SmSGs, and SlSGs (r = - 0.19, p = 0.03; r = - 0.02, p = 0.81; r = 0.12, p = 0.17, respectively). [11C]methionine uptake in the tori tubarius was correlated with that in the SmSGs and SlSGs (r = 0.24, p = 0.01; r = 0.32, p < 0.01, respectively), but not with that in the PSGs (r = 0.16, p = 0.08). CONCLUSIONS The TGs were undetectable on [99mTc]pertechnetate SPECT/CT. Both [18F]FDG and [11C]methionine uptakes in the tori tubarius were clearly affected by that in the palatine tonsils and was little related to that in the other salivary glands. Therefore, it seems difficult to evaluate the physiological function of the TGs as salivary glands using [99mTc]pertechnetate SPECT/CT, [18F]FDG PET/CT, and [11C]methionine PET/CT imaging.
Collapse
Affiliation(s)
- Yohji Matsusaka
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan.
| | - Tomohiko Yamane
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Kenji Fukushima
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Akira Seto
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Ichiro Matsunari
- Division of Nuclear Medicine, Department of Radiology, Saitama Medical University Hospital, 38 Moro-Hongo, Moroyama, 350-0495, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| |
Collapse
|
36
|
De la Vieja A, Riesco-Eizaguirre G. Radio-Iodide Treatment: From Molecular Aspects to the Clinical View. Cancers (Basel) 2021; 13:cancers13050995. [PMID: 33673669 PMCID: PMC7957486 DOI: 10.3390/cancers13050995] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary This year marks the 80th commemoration of the first time that radio-iodide treatment (RAI) was used. RAI is one of the most effective targeted internal radiation anticancer therapies ever devised and it has been used for many decades, however, a thorough understanding of the underlying molecular mechanisms involved could greatly improve the success of this therapy. This is an in-depth innovative review focusing on the molecular mechanisms underlying radio-iodide therapy in thyroid cancer and how the alteration of these mechanisms affects the results in the clinic. Abstract Thyroid radio-iodide therapy (RAI) is one of the oldest known and used targeted therapies. In thyroid cancer, it has been used for more than eight decades and is still being used to improve thyroid tumor treatment to eliminate remnants after thyroid surgery, and tumor metastases. Knowledge at the molecular level of the genes/proteins involved in the process has led to improvements in therapy, both from the point of view of when, how much, and how to use the therapy according to tumor type. The effectiveness of this therapy has spread into other types of targeted therapies, and this has made sodium/iodide symporter (NIS) one of the favorite theragnostic tools. Here we focus on describing the molecular mechanisms involved in radio-iodide therapy and how the alteration of these mechanisms in thyroid tumor progression affects the diagnosis and results of therapy in the clinic. We analyze basic questions when facing treatment, such as: (1) how the incorporation of radioiodine in normal, tumor, and metastatic thyroid cells occurs and how it is regulated; (2) the pros and cons of thyroid hormonal deprivation vs. recombinant human Thyroid Stimulating Hormone (rhTSH) in radioiodine residence time, treatment efficacy, thyroglobulin levels and organification, and its influence on diagnostic imaging tests and metastasis treatment; and (3) the effect of stunning and the possible causes. We discuss the possible incorporation of massive sequencing data into clinical practice, and we conclude with a socioeconomical and clinical vision of the above aspects.
Collapse
Affiliation(s)
- Antonio De la Vieja
- Endocrine Tumors Unit (Unidad Funcional de Investigación en Enfermedades Endocrinas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Correspondence: ; Tel.: +34-918223270
| | - Garcilaso Riesco-Eizaguirre
- Departamento de Endocrinología y Nutrición, Hospital Universitario de Móstoles, 28935 Madrid, Spain
- Molecular Endocrinology Group, Faculty of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| |
Collapse
|
37
|
Altmann L, Zantop V, Wenisch P, Diesendorf N, Heinrich MR. Visible Light Promoted, Catalyst-Free Radical Carbohydroxylation and Carboetherification under Mild Biomimetic Conditions. Chemistry 2021; 27:2452-2462. [PMID: 33006177 PMCID: PMC7898656 DOI: 10.1002/chem.202004234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Indexed: 11/25/2022]
Abstract
Metal and catalyst-free carbohydroxylations and carboetherifications at room temperature have been achieved by a combination of beneficial factors including high aryl diazonium concentration and visible light irradiation. The acceleration of the reaction by visible light irradiation is particularly remarkable against the background that neither the aryldiazonium salt nor the alkene show absorptions in the respective range of wavelength. These observations point to weak charge transfer interactions between diazonium salt and alkene, which are nevertheless able to considerably influence the reaction course. As highly promising perspective, many more aryldiazonium-based radical arylations might benefit from simple light irradiation without requiring a photocatalyst or particular additive.
Collapse
Affiliation(s)
- Lisa‐Marie Altmann
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Viviane Zantop
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Pia Wenisch
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Nina Diesendorf
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Markus R. Heinrich
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| |
Collapse
|
38
|
Dittmann M, Gonzalez Carvalho JM, Rahbar K, Schäfers M, Claesener M, Riemann B, Seifert R. Incremental diagnostic value of [ 18F]tetrafluoroborate PET-CT compared to [ 131I]iodine scintigraphy in recurrent differentiated thyroid cancer. Eur J Nucl Med Mol Imaging 2020; 47:2639-2646. [PMID: 32248325 PMCID: PMC7515952 DOI: 10.1007/s00259-020-04727-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/14/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Efficient therapy of recurrent differentiated thyroid cancer (DTC) is dependent on precise molecular imaging techniques targeting the human sodium iodide symporter (hNIS), which is a marker both of thyroid and DTC cells. Various iodine isotopes have been utilized for detecting DTC; however, these come with unfavorable radiation exposure and image quality ([131I]iodine) or limited availability ([124I]iodine). In contrast, [18F]tetrafluoroborate (TFB) is a novel radiolabeled PET substrate of hNIS, results in PET images with high-quality and low radiation doses, and should therefore be suited for imaging of DTC. The aim of the present study was to compare the diagnostic performance of [18F]TFB-PET to the clinical reference standard [131I]iodine scintigraphy in patients with recurrent DTC. METHODS Twenty-five patients with recurrent DTC were included in this retrospective analysis. All patients underwent [18F]TFB-PET combined with either CT or MRI due to newly discovered elevated TG levels, antiTG levels, sonographically suspicious cervical lymph nodes, or combinations of these findings. Correlative [131I]iodine whole-body scintigraphy (dxWBS) including SPECT-CT was present for all patients; correlative [18F]FDG-PET-CT was present for 21 patients. Histological verification of [18F]TFB positive findings was available in 4 patients. RESULTS [18F]TFB-PET detected local recurrence or metastases of DTC in significantly more patients than conventional [131I]iodine dxWBS and SPECT-CT (13/25 = 52% vs. 3/25 = 12%, p = 0.002). The diagnosis of 6 patients with cervical lymph node metastases that showed mildly increased FDG metabolism but negative [131I]iodine scintigraphy was changed: [18F]TFB-PET revealed hNIS expression in the metastases, which were therefore reclassified as only partly de-differentiated (histological confirmation present in two patients). Highest sensitivity for detecting recurrent DTC had the combination of [18F]TFB-PET-CT/MRI with [18F]FDG-PET-CT (64%). CONCLUSION In the present cohort, [18F]TFB-PET shows higher sensitivity and accuracy than [131I]iodine WBS and SPECT-CT in detecting recurrent DTC. The combination of [18F]TFB-PET with [18F]FDG-PET-CT seems a reasonable strategy to characterize DTC tumor manifestations with respect to their differentiation and thereby also individually plan and monitor treatment. Future prospective studies evaluating the potential of [18F]TFB-PET in recurrent DTC are warranted.
Collapse
Affiliation(s)
- Matthias Dittmann
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | | | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
- Cells in Motion Interfaculty Centre (CiM), University of Münster, Münster, Germany
| | - Michael Claesener
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Burkhard Riemann
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.
| |
Collapse
|
39
|
Concilio SC, Suksanpaisan L, Pham L, Peng KW, Russell SJ. Improved Noninvasive In Vivo Tracking of AAV-9 Gene Therapy Using the Perchlorate-Resistant Sodium Iodide Symporter from Minke Whale. Mol Ther 2020; 29:236-243. [PMID: 33038323 PMCID: PMC7791078 DOI: 10.1016/j.ymthe.2020.09.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 01/12/2023] Open
Abstract
The sodium iodide symporter (NIS) is widely used as a reporter gene to noninvasively monitor the biodistribution and durability of vector-mediated gene expression via gamma scintigraphy, single-photon emission computed tomography (SPECT), and positron-emission tomography (PET). However, the approach is limited by background signal due to radiotracer uptake by endogenous NIS-expressing tissues. In this study, using the SPECT tracer pertechnetate (99mTcO4) and the PET tracer tetrafluoroborate (B18F4), in combination with the NIS inhibitor perchlorate, we compared the transport properties of human NIS and minke whale (Balaenoptera acutorostrata scammoni) NIS in vitro and in vivo. Based on its relative resistance to perchlorate, the NIS protein from minke whale appeared to be the superior candidate reporter gene. SPECT and PET imaging studies in nude mice challenged with NIS-encoding adeno-associated virus (AAV)-9 vectors confirmed that minke whale NIS, in contrast to human and endogenous mouse NIS, continues to function as a reliable reporter even when background radiotracer uptake by endogenous NIS is blocked by perchlorate.
Collapse
Affiliation(s)
- Susanna C Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Linh Pham
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA.
| |
Collapse
|
40
|
Biermann M. [ 18F]tetrafluoroborate-PET Outperforms 131I-SPECT/CT in a Series of 25 Patients with Suspected Recurrent Differentiated Thyroid Cancer. Thyroid 2020. [DOI: 10.1089/ct.2020;32.394-398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Martin Biermann
- Nuclear Medicine/PET Center, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
41
|
Tutter M, Schug C, Schmohl KA, Urnauer S, Schwenk N, Petrini M, Lokerse WJM, Zach C, Ziegler S, Bartenstein P, Weber WA, Wagner E, Lindner LH, Nelson PJ, Spitzweg C. Effective control of tumor growth through spatial and temporal control of theranostic sodium iodide symporter ( NIS) gene expression using a heat-inducible gene promoter in engineered mesenchymal stem cells. Am J Cancer Res 2020; 10:4490-4506. [PMID: 32292510 PMCID: PMC7150485 DOI: 10.7150/thno.41489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/11/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose: The tumor homing characteristics of mesenchymal stem cells (MSCs) make them attractive vehicles for the tumor-specific delivery of therapeutic agents, such as the sodium iodide symporter (NIS). NIS is a theranostic protein that allows non-invasive monitoring of the in vivo biodistribution of functional NIS expression by radioiodine imaging as well as the therapeutic application of 131I. To gain local and temporal control of transgene expression, and thereby improve tumor selectivity, we engineered MSCs to express the NIS gene under control of a heat-inducible HSP70B promoter (HSP70B-NIS-MSCs). Experimental Design: NIS induction in heat-treated HSP70B-NIS-MSCs was verified by 125I uptake assay, RT-PCR, Western blot and immunofluorescence staining. HSP70B-NIS-MSCs were then injected i.v. into mice carrying subcutaneous hepatocellular carcinoma HuH7 xenografts, and hyperthermia (1 h at 41°C) was locally applied to the tumor. 0 - 72 h later radioiodine uptake was assessed by 123I-scintigraphy. The most effective uptake regime was then selected for 131I therapy. Results: The HSP70B promoter showed low basal activity in vitro and was significantly induced in response to heat. In vivo, the highest tumoral iodine accumulation was seen 12 h after application of hyperthermia. HSP70B-NIS-MSC-mediated 131I therapy combined with hyperthermia resulted in a significantly reduced tumor growth with prolonged survival as compared to control groups. Conclusions: The heat-inducible HSP70B promoter allows hyperthermia-induced spatial and temporal control of MSC-mediated theranostic NIS gene radiotherapy with efficient tumor-selective and temperature-dependent accumulation of radioiodine in heat-treated tumors.
Collapse
|
42
|
Zhekova HR, Sakuma T, Johnson R, Concilio SC, Lech PJ, Zdravkovic I, Damergi M, Suksanpaisan L, Peng KW, Russell SJ, Noskov S. Mapping of Ion and Substrate Binding Sites in Human Sodium Iodide Symporter (hNIS). J Chem Inf Model 2020; 60:1652-1665. [PMID: 32134653 DOI: 10.1021/acs.jcim.9b01114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The human sodium iodide symporter (hNIS) is a theranostic reporter gene which concentrates several clinically approved SPECT and PET radiotracers and plays an essential role for the synthesis of thyroid hormones as an iodide transporter in the thyroid gland. Development of hNIS mutants which could enhance translocation of the desired imaging ions is currently underway. Unfortunately, it is hindered by lack of understanding of the 3D organization of hNIS and its relation to anion transport. There are no known crystal structures of hNIS in any of its conformational states. Homology modeling can be very effective in such situations; however, the low sequence identity between hNIS and relevant secondary transporters with available experimental structures makes the choice of a template and the generation of 3D models nontrivial. Here, we report a combined application of homology modeling and molecular dynamics refining of the hNIS structure in its semioccluded state. The modeling was based on templates from the LeuT-fold protein family and was done with emphasis on the refinement of the substrate-ion binding pocket. The consensus model developed in this work is compared to available biophysical and biochemical experimental data for a number of different LeuT-fold proteins. Some functionally important residues contributing to the formation of putative binding sites and permeation pathways for the cotransported Na+ ions and I- substrate were identified. The model predictions were experimentally tested by generation of mutant versions of hNIS and measurement of relative (to WT hNIS) 125I- uptake of 35 hNIS variants.
Collapse
Affiliation(s)
- Hristina R Zhekova
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Toshie Sakuma
- Imanis Life Sciences, Rochester, Minnesota 55901, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Ryan Johnson
- Imanis Life Sciences, Rochester, Minnesota 55901, United States
| | - Susanna C Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Patrycja J Lech
- Imanis Life Sciences, Rochester, Minnesota 55901, United States
| | - Igor Zdravkovic
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mirna Damergi
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Kah-Whye Peng
- Imanis Life Sciences, Rochester, Minnesota 55901, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Stephen J Russell
- Imanis Life Sciences, Rochester, Minnesota 55901, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Sergei Noskov
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
43
|
Concilio SC, Zhekova HR, Noskov SY, Russell SJ. Inter-species variation in monovalent anion substrate selectivity and inhibitor sensitivity in the sodium iodide symporter (NIS). PLoS One 2020; 15:e0229085. [PMID: 32084174 PMCID: PMC7034854 DOI: 10.1371/journal.pone.0229085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
The sodium iodide symporter (NIS) transports iodide, which is necessary for thyroid hormone production. NIS also transports other monovalent anions such as tetrafluoroborate (BF4-), pertechnetate (TcO4-), and thiocyanate (SCN-), and is competitively inhibited by perchlorate (ClO4-). However, the mechanisms of substrate selectivity and inhibitor sensitivity are poorly understood. Here, a comparative approach was taken to determine whether naturally evolved NIS proteins exhibit variability in their substrate transport properties. The NIS proteins of thirteen animal species were initially assessed, and three species from environments with differing iodide availability, freshwater species Danio rerio (zebrafish), saltwater species Balaenoptera acutorostrata scammoni (minke whale), and non-aquatic mammalian species Homo sapiens (human) were studied in detail. NIS genes from each of these species were lentivirally transduced into HeLa cells, which were then characterized using radioisotope uptake assays, 125I- competitive substrate uptake assays, and kinetic assays. Homology models of human, minke whale and zebrafish NIS were used to evaluate sequence-dependent impact on the organization of Na+ and I- binding pockets. Whereas each of the three proteins that were analyzed in detail concentrated iodide to a similar degree, their sensitivity to perchlorate inhibition varied significantly: minke whale NIS was the least impacted by perchlorate inhibition (IC50 = 4.599 μM), zebrafish NIS was highly sensitive (IC50 = 0.081 μM), and human NIS showed intermediate sensitivity (IC50 = 1.566 μM). Further studies with fifteen additional substrates and inhibitors revealed similar patterns of iodide uptake inhibition, though the degree of 125I- uptake inhibition varied with each compound. Kinetic analysis revealed whale NIS had the lowest Km-I and the highest Vmax-I. Conversely, zebrafish NIS had the highest Km and lowest Vmax. Again, human NIS was intermediate. Molecular modeling revealed a high degree of conservation in the putative ion binding pockets of NIS proteins from different species, which suggests the residues responsible for the observed differences in substrate selectivity lie elsewhere in the protein. Ongoing studies are focusing on residues in the extracellular loops of NIS as determinants of anion specificity. These data demonstrate significant transport differences between the NIS proteins of different species, which may be influenced by the unique physiological needs of each organism. Our results also identify naturally-existing NIS proteins with significant variability in substrate transport kinetics and inhibitor sensitivity, which suggest that the affinity and selectivity of NIS for certain substrates can be altered for biotechnological and clinical applications. Further examination of interspecies differences may improve understanding of the substrate transport mechanism.
Collapse
Affiliation(s)
- Susanna C. Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hristina R. Zhekova
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Sergei Y. Noskov
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
44
|
Vail DM, LeBlanc AK, Jeraj R. Advanced Cancer Imaging Applied in the Comparative Setting. Front Oncol 2020; 10:84. [PMID: 32117739 PMCID: PMC7019008 DOI: 10.3389/fonc.2020.00084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 11/13/2022] Open
Abstract
The potential for companion (pet) species with spontaneously arising tumors to act as surrogates for preclinical development of advanced cancer imaging technologies has become more apparent in the last decade. The utility of the companion model specifically centers around issues related to body size (including spatial target/normal anatomic characteristics), physical size and spatial distribution of metastasis, tumor heterogeneity, the presence of an intact syngeneic immune system and a syngeneic tumor microenvironment shaped by the natural evolution of the cancer. Companion species size allows the use of similar equipment, hardware setup, software, and scan protocols which provide the opportunity for standardization and harmonization of imaging operating procedures and quality assurance across imaging protocols, imaging hardware, and the imaged species. Murine models generally do not replicate the size and spatial distribution of human metastatic cancer and these factors strongly influence image resolution and dosimetry. The following review will discuss several aspects of comparative cancer imaging in more detail while providing several illustrative examples of investigational approaches performed or currently under exploration at our institutions. Topics addressed include a discussion on interested consortia; image quality assurance and harmonization; image-based biomarker development and validation; contrast agent and radionuclide tracer development; advanced imaging to assess and predict response to cytotoxic and immunomodulatory anticancer agents; imaging of the tumor microenvironment; development of novel theranostic approaches; cell trafficking assessment via non-invasive imaging; and intraoperative imaging to inform surgical oncology decision making. Taken in totality, these comparative opportunities predict that safety, diagnostic and efficacy data generated in companion species with naturally developing and progressing cancers would better recapitulate the human cancer condition than that of artificial models in small rodent systems and ultimately accelerate the integration of novel imaging technologies into clinical practice. It is our hope that the examples presented should serve to provide those involved in cancer investigations who are unfamiliar with available comparative methodologies an understanding of the potential utility of this approach.
Collapse
Affiliation(s)
- David M Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Robert Jeraj
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
45
|
Okamura T, Tsukamoto S, Nagatsu K, Okada M, Minegishi K, Tatsumi T, Sugyo A, Kikuchi T, Wakizaka H, Ishii H, Tsuji AB, Zhang MR. 6-[124I]Iodo-9-pentylpurine for Imaging the Activity of the Sodium Iodide Symporter in the Brain. J Med Chem 2020; 63:1717-1723. [DOI: 10.1021/acs.jmedchem.9b02096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Toshimitsu Okamura
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Satoshi Tsukamoto
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kotaro Nagatsu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Maki Okada
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Katsuyuki Minegishi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Takayuki Tatsumi
- Comprehensive Reproductive Medicine, Regulation of Internal Environment and Reproduction, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Aya Sugyo
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tatsuya Kikuchi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hidekatsu Wakizaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hideki Ishii
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Atsushi B. Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
46
|
Punzón I, Mauduit D, Holvoet B, Thibaud JL, de Fornel P, Deroose CM, Blanchard-Gutton N, Vilquin JT, Sampaolesi M, Barthélémy I, Blot S. In Vivo Myoblasts Tracking Using the Sodium Iodide Symporter Gene Expression in Dogs. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:317-327. [PMID: 32577429 PMCID: PMC7293195 DOI: 10.1016/j.omtm.2019.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 12/13/2019] [Indexed: 01/07/2023]
Abstract
Stem cell-based therapies are a promising approach for the treatment of degenerative muscular diseases; however, clinical trials have shown inconclusive and even disappointing results so far. Noninvasive cell monitoring by medicine imaging could improve the understanding of the survival and biodistribution of cells following injection. In this study, we assessed the canine sodium iodide symporter (cNIS) reporter gene as an imaging tool to track by single-photon emission computed tomography (SPECT/CT) transduced canine myoblasts after intramuscular (IM) administrations in dogs. cNIS-expressing cells kept their myogenic capacities and showed strong 99 mTc-pertechnetate (99 mTcO4−) uptake efficiency both in vitro and in vivo. cNIS expression allowed visualization of cells by SPECT/CT along time: 4 h, 48 h, 7 days, and 30 days after IM injection; biopsies collected 30 days post administration showed myofiber’s membranes expressing cNIS. This study demonstrates that NIS can be used as a reporter to track cells in vivo in the skeletal muscle of large animals. Our results set a proof of concept of the benefits NIS-tracking tool may bring to the already challenging cell-based therapies arena in myopathies and pave the way to a more efficient translation to the clinical setting from more accurate pre-clinical results.
Collapse
Affiliation(s)
- Isabel Punzón
- INSERM U955-E10, IMRB, Université Paris Est Créteil, Ecole nationale vétérinaire d'Alfort, 94700 Maisons-Alfort, France
| | - David Mauduit
- INSERM U955-E10, IMRB, Université Paris Est Créteil, Ecole nationale vétérinaire d'Alfort, 94700 Maisons-Alfort, France
| | - Bryan Holvoet
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | | | | | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Nicolas Blanchard-Gutton
- INSERM U955-E10, IMRB, Université Paris Est Créteil, Ecole nationale vétérinaire d'Alfort, 94700 Maisons-Alfort, France
| | - Jean-Thomas Vilquin
- Sorbonne Université, INSERM, AIM, Centre de Recherche en Myologie, UMRS 974, AP-HP, Hôpital Pitié Salpêtrière, 75013 Paris, France
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Inès Barthélémy
- INSERM U955-E10, IMRB, Université Paris Est Créteil, Ecole nationale vétérinaire d'Alfort, 94700 Maisons-Alfort, France
| | - Stéphane Blot
- INSERM U955-E10, IMRB, Université Paris Est Créteil, Ecole nationale vétérinaire d'Alfort, 94700 Maisons-Alfort, France
| |
Collapse
|
47
|
Zhang L, Suksanpaisan L, Jiang H, DeGrado TR, Russell SJ, Zhao M, Peng KW. Dual-Isotope SPECT Imaging with NIS Reporter Gene and Duramycin to Visualize Tumor Susceptibility to Oncolytic Virus Infection. Mol Ther Oncolytics 2019; 15:178-185. [PMID: 31890867 PMCID: PMC6931109 DOI: 10.1016/j.omto.2019.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/05/2019] [Indexed: 11/21/2022] Open
Abstract
Noninvasive dual-imaging methods that provide an early readout on tumor permissiveness to virus infection and tumor cell death could be valuable in optimizing development of oncolytic virotherapies. Here, we have used the sodium iodide symporter (NIS) and 125I radiotracer to detect infection and replicative spread of an oncolytic vesicular stomatitis virus (VSV) in VSV-susceptible (MPC-11 tumor) versus VSV-resistant (CT26 tumor) tumors in BALB/c mice. In conjunction, tumor cell death was imaged simultaneously using technetium (99mTc)-duramycin that binds phosphatidylethanolamine in apoptotic and necrotic cells. Dual-isotope single-photon emission computed tomography/computed tomography (SPECT/CT) imaging showed areas of virus infection (NIS and 125I), which overlapped well with areas of tumor cell death (99mTc-duramycin imaging) in susceptible tumors. Multiple infectious foci arose early in MPC-11 tumors, which rapidly expanded throughout the tumor parenchyma over time. There was a dose-dependent increase in numbers of infectious centers and 99mTc-duramycin-positive areas with viral dose. In contrast, NIS or duramycin signals were minimal in VSV-resistant CT26 tumors. Combinatorial use of NIS and 99mTc-duramycin SPECT imaging for simultaneous monitoring of oncolytic virotherapy (OV) spread and the presence or absence of treatment-associated cell death could be useful to guide development of combination treatment strategies to enhance therapeutic outcome.
Collapse
Affiliation(s)
- Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Huailei Jiang
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Ming Zhao
- Northwestern University, Chicago, IL, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
48
|
Burke BP, Grantham W, Burke MJ, Nichol GS, Roberts D, Renard I, Hargreaves R, Cawthorne C, Archibald SJ, Lusby PJ. Visualizing Kinetically Robust Co III4L 6 Assemblies in Vivo: SPECT Imaging of the Encapsulated [ 99mTc]TcO 4- Anion. J Am Chem Soc 2018; 140:16877-16881. [PMID: 30485075 DOI: 10.1021/jacs.8b09582] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Noncovalent encapsulation is an attractive approach for modifying the efficacy and physiochemical properties of both therapeutic and diagnostic species. Abiotic self-assembled constructs have shown promise, yet many hurdles between in vitro and (pre)clinical studies remain, not least the challenges associated with maintaining the macromolecular, hollow structure under nonequilibrium conditions. Using a kinetically robust CoIII4L6 tetrahedron we now show the feasibility of encapsulating the most widely used precursor in clinical nuclear diagnostic imaging, the γ-emitting [99mTc]TcO4- anion, under conditions compatible with in vivo administration. Subsequent single-photon emission computed tomography imaging of the caged-anion reveals a marked change in the biodistribution compared to the thyroid-accumulating free oxo-anion, thus moving clinical applications of (metallo)supramolecular species a step closer.
Collapse
Affiliation(s)
- Benjamin P Burke
- Department of Chemistry , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom.,Positron Emission Tomography Research Centre , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom
| | - William Grantham
- EaStCHEM School of Chemistry , University of Edinburgh , Joseph Black Building, David Brewster Road , Edinburgh EH9 3FJ , Scotland
| | - Michael J Burke
- EaStCHEM School of Chemistry , University of Edinburgh , Joseph Black Building, David Brewster Road , Edinburgh EH9 3FJ , Scotland
| | - Gary S Nichol
- EaStCHEM School of Chemistry , University of Edinburgh , Joseph Black Building, David Brewster Road , Edinburgh EH9 3FJ , Scotland
| | - David Roberts
- School of Life Sciences , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom.,Positron Emission Tomography Research Centre , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom
| | - Isaline Renard
- Department of Chemistry , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom.,Positron Emission Tomography Research Centre , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom
| | - Rebecca Hargreaves
- Department of Chemistry , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom.,Positron Emission Tomography Research Centre , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom
| | - Christopher Cawthorne
- School of Life Sciences , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom.,Positron Emission Tomography Research Centre , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom
| | - Stephen J Archibald
- Department of Chemistry , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom.,Positron Emission Tomography Research Centre , University of Hull , Cottingham Road , Hull HU6 7RX , United Kingdom
| | - Paul J Lusby
- EaStCHEM School of Chemistry , University of Edinburgh , Joseph Black Building, David Brewster Road , Edinburgh EH9 3FJ , Scotland
| |
Collapse
|
49
|
Cui Q, Liu Y, Zhou M, Han Y, Yin C, Bai G. An Optimized MicroPET Imaging Method for the Distribution and Synergies of Natural Products. Front Pharmacol 2018; 9:948. [PMID: 30186178 PMCID: PMC6110851 DOI: 10.3389/fphar.2018.00948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/02/2018] [Indexed: 12/03/2022] Open
Abstract
Purpose: Understanding the distribution and interaction of the Traditional Chinese Medicines (TCMs) is an integral source of herbal drug discovery. An optimized radio-labeled method was explored that could conduct in situ biodistribution studies in animals. We evaluated the feasibility of the method and applied glycyrrhetinic acid and platycodon (PG) polysaccharides as models. Procedures: [18F]-GA is a novel radiotracer which was performed positron emission tomography (PET) studies to assay the biodistribution of GA in mice. In addition, PG polysaccharide was used to intervene the biodistribution and dosimetry of GA. Scanning data were analyzed with professional software. Results: Record the time-activity curves for all organs then use the normalization method to calculate the area under the curve as a dosimetry for each organ. Moreover, the addition of PG polysaccharides can significantly improve the dosimetry of GA in the lungs, and its effect was related to the administration time. Conclusion: MicroPET imaging opens up a new avenue for the application of drug interactions between the TCMs.
Collapse
Affiliation(s)
- Qingxin Cui
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yang Liu
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Mengge Zhou
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yanqi Han
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Chengcheng Yin
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Gang Bai
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|