1
|
Costa IM, Firth G, Kim J, Banu A, Pham TT, Sunassee K, Langdon S, De Santis V, Vass L, Schettino G, Fruhwirth GO, Terry SYA. In Vitro and Preclinical Systematic Dose-Effect Studies of Auger Electron- and β Particle-Emitting Radionuclides and External Beam Radiation for Cancer Treatment. Int J Radiat Oncol Biol Phys 2024; 120:1124-1134. [PMID: 38797497 PMCID: PMC7616868 DOI: 10.1016/j.ijrobp.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/09/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Despite a rise in clinical use of radiopharmaceutical therapies, the biological effects of radionuclides and their relationship with absorbed radiation dose are poorly understood. Here, we set out to define this relationship for Auger electron emitters [99mTc]TcO4- and [123I]I- and β--particle emitter [188Re]ReO4-. Studies were carried out using genetically modified cells that permitted direct radionuclide comparisons. METHODS AND MATERIALS Triple-negative MDA-MB-231 breast cancer cells expressing the human sodium iodide symporter (hNIS) and green fluorescent protein (GFP; MDA-MB-231.hNIS-GFP) were used. In vitro radiotoxicity of [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- was determined using clonogenic assays. Radionuclide uptake, efflux, and subcellular location were used to calculate nuclear absorbed doses using the Medical Internal Radiation Dose (MIRD) formalism. In vivo studies were performed using female NSG mice bearing orthotopic MDA-MB-231.hNIS-GFP tumors and compared with X-ray-treated (12.6-15 Gy) and untreated cohorts. Absorbed dose per unit activity in tumors and sodium iodide symporter-expressing organs was extrapolated to reference human adult models using OLINDA/EXM. RESULTS [99mTc]TcO4- and [123I]I- reduced the survival fraction only in hNIS-expressing cells, whereas [188Re]ReO4- reduced survival fraction in hNIS-expressing and parental cells. [123I]I- required 2.4- and 1.5-fold lower decays/cell to achieve 37% survival compared with [99mTc]TcO4- and [188Re]ReO4-, respectively, after 72 hours of incubation. Additionally, [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- had superior cell killing effectiveness in vitro compared with X-rays. In vivo, X-ray led to a greater median survival compared with [188Re]ReO4- and [123I]I- (54 days vs 45 and 43 days, respectively). Unlike the X-ray cohort, no metastases were visualized in the radionuclide-treated cohorts. Extrapolated human absorbed doses of [188Re]ReO4- to a 1 g tumor were 13.8- and 11.2-fold greater than for [123I]I- in female and male models, respectively. CONCLUSIONS This work reports reference dose-effect data using cell and tumor models for [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- for the first time. We further demonstrate the tumor-controlling effects of [123I]I- and [188Re]ReO4- in comparison with external beam radiation therapy.
Collapse
Affiliation(s)
- Ines M Costa
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom; Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - George Firth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Jana Kim
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Arshiya Banu
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Truc T Pham
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Kavitha Sunassee
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sophie Langdon
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Vittorio De Santis
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Laurence Vass
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Giuseppe Schettino
- Medical Radiation Science Group, National Physical Laboratory, Teddington, United Kingdom.
| | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom.
| | - Samantha Y A Terry
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
2
|
Musket A, Davern S, Elam BM, Musich PR, Moorman JP, Jiang Y. The application of radionuclide therapy for breast cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 3:1323514. [PMID: 39355029 PMCID: PMC11440853 DOI: 10.3389/fnume.2023.1323514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/27/2023] [Indexed: 10/03/2024]
Abstract
Radionuclide-mediated diagnosis and therapy have emerged as effective and low-risk approaches to treating breast cancer. Compared to traditional anatomic imaging techniques, diagnostic radionuclide-based molecular imaging systems exhibit much greater sensitivity and ability to precisely illustrate the biodistribution and metabolic processes from a functional perspective in breast cancer; this transitions diagnosis from an invasive visualization to a noninvasive visualization, potentially ensuring earlier diagnosis and on-time treatment. Radionuclide therapy is a newly developed modality for the treatment of breast cancer in which radionuclides are delivered to tumors and/or tumor-associated targets either directly or using delivery vehicles. Radionuclide therapy has been proven to be eminently effective and to exhibit low toxicity when eliminating both primary tumors and metastases and even undetected tumors. In addition, the specific interaction between the surface modules of the delivery vehicles and the targets on the surface of tumor cells enables radionuclide targeting therapy, and this represents an exceptional potential for this treatment in breast cancer. This article reviews the development of radionuclide molecular imaging techniques that are currently employed for early breast cancer diagnosis and both the progress and challenges of radionuclide therapy employed in breast cancer treatment.
Collapse
Affiliation(s)
- Anna Musket
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Sandra Davern
- Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Brianna M Elam
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Philip R Musich
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Jonathan P Moorman
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yong Jiang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
3
|
Belyaev IB, Zelepukin IV, Tishchenko VK, Petriev VM, Trushina DB, Klimentov SM, Zavestovskaya IN, Ivanov SA, Kaprin AD, Deyev SM, Kabashin AV. Nanoparticles based on MIL-101 metal-organic frameworks as efficient carriers of therapeutic 188Re radionuclide for nuclear medicine. NANOTECHNOLOGY 2023; 35:075103. [PMID: 37963406 DOI: 10.1088/1361-6528/ad0c74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/14/2023] [Indexed: 11/16/2023]
Abstract
Nuclear medicine presents one of the most promising modalities for efficient non-invasive treatment of a variety of cancers, but the application of radionuclides in cancer therapy and diagnostics is severely limited by their nonspecific tissue accumulation and poor biocompatibility. Here, we explore the use of nanosized metal-organic frameworks (MOFs) as carriers of radionuclides to order to improve their delivery to tumour. To demonstrate the concept, we prepared polymer-coated MIL-101(Cr)-NH2MOFs and conjugated them with clinically utilized radionuclide188Re. The nanoparticles demonstrated high loading efficacy of radionuclide reaching specific activity of 49 MBq mg-1. Pharmacokinetics of loaded MOFs was investigated in mice bearing colon adenocarcinoma. The biological half-life of the radionuclide in blood was (20.9 ± 1.3) h, and nanoparticles enabled it to passively accumulate and retain in the tumour. The radionuclide delivery with MOFs led to a significant decrease of radioactivity uptake by the thyroid gland and stomach as compared with perrhenate salt injection, which is beneficial for reducing the side toxicity of nuclear therapy. The reported data on the functionalization and pharmacokinetics of MIL-101(Cr)-NH2for radionuclide delivery unveils the promising potential of these MOFs for nuclear medicine.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ivan V Zelepukin
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Victoria K Tishchenko
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Vasiliy M Petriev
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Daria B Trushina
- Federal Research Center 'Crystallography and Photonics', Russian Academy of Sciences, Moscow 119333, Russia
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Sergey M Klimentov
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
| | - Irina N Zavestovskaya
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
| | - Sergey A Ivanov
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Andrey D Kaprin
- A. Tsyb Medical Radiological Research Centre, Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia
| | - Sergey M Deyev
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Institute for Physics and Engineering in Biomedicine (PhysBio), Moscow 115409, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
- Kazan Federal University, Institute of Fundamental Medicine and Biology, 420008, Kazan, Russia
| | - Andrei V Kabashin
- Aix Marseille University, CNRS, LP3, Campus de Luminy, Case 917, F-13288, Marseille, France
| |
Collapse
|
4
|
Fundamentals of Rhenium-188 Radiopharmaceutical Chemistry. Molecules 2023; 28:molecules28031487. [PMID: 36771153 PMCID: PMC9921938 DOI: 10.3390/molecules28031487] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The β- emitter, rhenium-188 (188Re), has long been recognized as an attractive candidate for targeted cancer radionuclide therapy (TRNT). This transition metal shares chemical similarities with its congener element technetium, whose nuclear isomer technetium-99m (99mTc) is the current workhorse of diagnostic nuclear medicine. The differences between these two elements have a significant impact on the radiolabelling methods and should always receive critical attention. This review aims to highlight what needs to be considered to design a successful radiopharmaceutical incorporating 118Re. Some of the most effective strategies for preparing therapeutic radiopharmaceuticals with 188Re are illustrated and rationalized using the concept of the inorganic functional group (core) and a simple ligand field theoretical model combined with a qualitative definition of frontiers orbitals. Of special interest are the Re(V) oxo and Re(V) nitrido functional groups. Suitable ligands for binding to these cores are discussed, successful clinical applications are summarized, and a prediction of viable future applications is presented. Rhenium-188 decays through the emission of a high energy beta particle (2.12 MeV max energy) and a half-life of 16.9 h. An ideal biological target would therefore be a high-capacity target site (transporters, potential gradients, tumour microenvironment) with less emphasis on saturable targets such as overexpressed receptors on smaller metastases.
Collapse
|
5
|
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: 11] [Impact Index Per Article: 3.7] [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
|
6
|
Paquet F, Leggett RW, Blanchardon E, Bailey MR, Gregoratto D, Smith T, Ratia G, Davesne E, Berkovski V, Harrison JD. Occupational Intakes of Radionuclides: Part 5. Ann ICRP 2022; 51:11-415. [PMID: 35414227 DOI: 10.1177/01466453211028755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
7
|
Shalaby N, Dubois VP, Ronald J. Molecular imaging of cellular immunotherapies in experimental and therapeutic settings. Cancer Immunol Immunother 2021; 71:1281-1294. [PMID: 34657195 PMCID: PMC9122865 DOI: 10.1007/s00262-021-03073-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 09/28/2021] [Indexed: 11/27/2022]
Abstract
Cell-based cancer immunotherapies are becoming a routine part of the armamentarium against cancer. While remarkable successes have been seen, including durable remissions, not all patients will benefit from these therapies and many can suffer from life-threatening side effects. These differences in efficacy and safety across patients and across tumor types (e.g., blood vs. solid), are thought to be due to differences in how well the immune cells traffic to their target tissue (e.g., tumor, lymph nodes, etc.) whilst avoiding non-target tissues. Across patient variability can also stem from whether the cells interact with (i.e., communicate with) their intended target cells (e.g., cancer cells), as well as if they proliferate and survive long enough to yield potent and long-lasting therapeutic effects. However, many cell-based therapies are monitored by relatively simple blood tests that lack any spatial information and do not reflect how many immune cells have ended up at particular tissues. The ex vivo labeling and imaging of infused therapeutic immune cells can provide a more precise and dynamic understanding of whole-body immune cell biodistribution, expansion, viability, and activation status in individual patients. In recent years numerous cellular imaging technologies have been developed that may provide this much-needed information on immune cell fate. For this review, we summarize various ex vivo labeling and imaging approaches that allow for tracking of cellular immunotherapies for cancer. Our focus is on clinical imaging modalities and summarize the progression from experimental to therapeutic settings. The imaging information provided by these technologies can potentially be used for many purposes including improved real-time understanding of therapeutic efficacy and potential side effects in individual patients after cell infusion; the ability to more readily compare new therapeutic cell designs to current designs for various parameters such as improved trafficking to target tissues and avoidance of non-target tissues; and the long-term ability to identify patient populations that are likely to be positive responders and at low-risk of side effects.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.,Robarts Research Institute, London, Ontario, Canada
| | - Veronica Phyllis Dubois
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.,Robarts Research Institute, London, Ontario, Canada
| | - John Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada. .,Robarts Research Institute, London, Ontario, Canada. .,Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
8
|
Pellico J, Gawne PJ, T M de Rosales R. Radiolabelling of nanomaterials for medical imaging and therapy. Chem Soc Rev 2021; 50:3355-3423. [PMID: 33491714 DOI: 10.1039/d0cs00384k] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanomaterials offer unique physical, chemical and biological properties of interest for medical imaging and therapy. Over the last two decades, there has been an increasing effort to translate nanomaterial-based medicinal products (so-called nanomedicines) into clinical practice and, although multiple nanoparticle-based formulations are clinically available, there is still a disparity between the number of pre-clinical products and those that reach clinical approval. To facilitate the efficient clinical translation of nanomedicinal-drugs, it is important to study their whole-body biodistribution and pharmacokinetics from the early stages of their development. Integrating this knowledge with that of their therapeutic profile and/or toxicity should provide a powerful combination to efficiently inform nanomedicine trials and allow early selection of the most promising candidates. In this context, radiolabelling nanomaterials allows whole-body and non-invasive in vivo tracking by the sensitive clinical imaging techniques positron emission tomography (PET), and single photon emission computed tomography (SPECT). Furthermore, certain radionuclides with specific nuclear emissions can elicit therapeutic effects by themselves, leading to radionuclide-based therapy. To ensure robust information during the development of nanomaterials for PET/SPECT imaging and/or radionuclide therapy, selection of the most appropriate radiolabelling method and knowledge of its limitations are critical. Different radiolabelling strategies are available depending on the type of material, the radionuclide and/or the final application. In this review we describe the different radiolabelling strategies currently available, with a critical vision over their advantages and disadvantages. The final aim is to review the most relevant and up-to-date knowledge available in this field, and support the efficient clinical translation of future nanomedicinal products for in vivo imaging and/or therapy.
Collapse
Affiliation(s)
- Juan Pellico
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK.
| | | | | |
Collapse
|
9
|
Asadian S, Mirzaei H, Kalantari BA, Davarpanah MR, Mohamadi M, Shpichka A, Nasehi L, Es HA, Timashev P, Najimi M, Gheibi N, Hassan M, Vosough M. β-radiating radionuclides in cancer treatment, novel insight into promising approach. Pharmacol Res 2020; 160:105070. [PMID: 32659429 DOI: 10.1016/j.phrs.2020.105070] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Targeted radionuclide therapy, known as molecular radiotherapy is a novel therapeutic module in cancer medicine. β-radiating radionuclides have definite impact on target cells via interference in cell cycle and particular signalings that can lead to tumor regression with minimal off-target effects on the surrounding tissues. Radionuclides play a remarkable role not only in apoptosis induction and cell cycle arrest, but also in the amelioration of other characteristics of cancer cells. Recently, application of novel β-radiating radionuclides in cancer therapy has been emerged as a promising therapeutic modality. Several investigations are ongoing to understand the underlying molecular mechanisms of β-radiating elements in cancer medicine. Based on the radiation dose, exposure time and type of the β-radiating element, different results could be achieved in cancer cells. It has been shown that β-radiating radioisotopes block cancer cell proliferation by inducing apoptosis and cell cycle arrest. However, physical characteristics of the β-radiating element (half-life, tissue penetration range, and maximum energy) and treatment protocol determine whether tumor cells undergo cell cycle arrest, apoptosis or both and to which extent. In this review, we highlighted novel therapeutic effects of β-radiating radionuclides on cancer cells, particularly apoptosis induction and cell cycle arrest.
Collapse
Affiliation(s)
- Samieh Asadian
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | | | - Morteza Mohamadi
- Department of Physical Chemistry, Faculty of Science, University of Tehran, Tehran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Leila Nasehi
- Department of Medical Laboratory Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; Department of Polymers and Composites, NN Semenov Institute of Chemical Physics, Moscow, Russia.
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research, Université Catholique de Louvain, B-1200 Brussels, Belgium
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
10
|
Klenner MA, Darwish T, Fraser BH, Massi M, Pascali G. Labeled Rhenium Complexes: Radiofluorination, α-MSH Cyclization, and Deuterium Substitutions. Organometallics 2020. [DOI: 10.1021/acs.organomet.0c00267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mitchell A. Klenner
- National Deuteration Facility (NDF) & Human Health, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales, Australia 2234
- School of Molecular and Life Sciences, Curtin University, Bentley, Western Australia, Australia 6102
| | - Tamim Darwish
- National Deuteration Facility (NDF) & Human Health, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales, Australia 2234
| | - Benjamin H. Fraser
- National Deuteration Facility (NDF) & Human Health, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales, Australia 2234
| | - Massimiliano Massi
- School of Molecular and Life Sciences, Curtin University, Bentley, Western Australia, Australia 6102
| | - Giancarlo Pascali
- National Deuteration Facility (NDF) & Human Health, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales, Australia 2234
- Prince of Wales Hospital, Randwick, New South Wales, Australia 2031
- School of Chemistry, University of New South Wales (UNSW), Kensington, New South Wales, Australia 2052
| |
Collapse
|
11
|
Jian J, Varathan E, Cheisson T, Jian T, Lukens WW, Davis RL, Schelter EJ, Schreckenbach G, Gibson JK. Proton affinities of pertechnetate (TcO 4-) and perrhenate (ReO 4-). Phys Chem Chem Phys 2020; 22:12403-12411. [PMID: 32452480 DOI: 10.1039/d0cp01721c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The anions pertechnetate, TcO4-, and perrhenate, ReO4-, exhibit very similar chemical and physical properties. Revealing and understanding disparities between them enhances fundamental understanding of both. Electrospray ionization generated the gas-phase proton bound dimer (TcO4-)(H+)(ReO4-). Collision induced dissociation of the dimer yielded predominantly HTcO4 and ReO4-, which according to Cooks' kinetic method indicates that the proton affinity (PA) of TcO4- is greater than that of ReO4-. Density functional theory computations agree with the experimental observation, providing PA[TcO4-] = 300.1 kcal mol-1 and PA[ReO4-] = 297.2 kcal mol-1. Attempts to rationalize these relative PAs based on elementary molecular parameters such as atomic charges indicate that the entirety of bond formation and concomitant bond disruption needs to be considered to understand the energies associated with such protonation processes. Although in both the gas and solution phases, TcO4- is a stronger base than ReO4-, it is noted that the significance of even such qualitative accordance is tempered by the very different natures of the underlying phenomena.
Collapse
Affiliation(s)
- Jiwen Jian
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
| | - Elumalai Varathan
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada.
| | - Thibault Cheisson
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Tian Jian
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
| | - Wayne W Lukens
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
| | - Rebecca L Davis
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada.
| | - Eric J Schelter
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Georg Schreckenbach
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada.
| | - John K Gibson
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
| |
Collapse
|
12
|
Lepareur N, Lacœuille F, Bouvry C, Hindré F, Garcion E, Chérel M, Noiret N, Garin E, Knapp FFR. Rhenium-188 Labeled Radiopharmaceuticals: Current Clinical Applications in Oncology and Promising Perspectives. Front Med (Lausanne) 2019; 6:132. [PMID: 31259173 PMCID: PMC6587137 DOI: 10.3389/fmed.2019.00132] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022] Open
Abstract
Rhenium-188 (188Re) is a high energy beta-emitting radioisotope with a short 16.9 h physical half-life, which has been shown to be a very attractive candidate for use in therapeutic nuclear medicine. The high beta emission has an average energy of 784 keV and a maximum energy of 2.12 MeV, sufficient to penetrate and destroy targeted abnormal tissues. In addition, the low-abundant gamma emission of 155 keV (15%) is efficient for imaging and for dosimetric calculations. These key characteristics identify 188Re as an important therapeutic radioisotope for routine clinical use. Moreover, the highly reproducible on-demand availability of 188Re from the 188W/188Re generator system is an important feature and permits installation in hospital-based or central radiopharmacies for cost-effective availability of no-carrier-added (NCA) 188Re. Rhenium-188 and technetium-99 m exhibit similar chemical properties and represent a "theranostic pair." Thus, preparation and targeting of 188Re agents for therapy is similar to imaging agents prepared with 99mTc, the most commonly used diagnostic radionuclide. Over the last three decades, radiopharmaceuticals based on 188Re-labeled small molecules, including peptides, antibodies, Lipiodol and particulates have been reported. The successful application of these 188Re-labeled therapeutic radiopharmaceuticals has been reported in multiple early phase clinical trials for the management of various primary tumors, bone metastasis, rheumatoid arthritis, and endocoronary interventions. This article reviews the use of 188Re-radiopharmaceuticals which have been investigated in patients for cancer treatment, demonstrating that 188Re represents a cost effective alternative for routine clinical use in comparison to more expensive and/or less readily available therapeutic radioisotopes.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène MarquisRennes, France
- Univ RennesInra, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer)—UMR_A 1341, UMR_S 1241, Rennes, France
| | - Franck Lacœuille
- Angers University HospitalAngers, France
- Univ AngersUniv Nantes, Inserm, CNRS, CRCINA (Centre de Recherche en Cancérologie et Immunologie Nantes—Angers)—UMR 1232, ERL 6001, Nantes, France
| | - Christelle Bouvry
- Comprehensive Cancer Center Eugène MarquisRennes, France
- Univ RennesCNRS, ISCR (Institut des Sciences Chimiques de Rennes)—UMR 6226, Rennes, France
| | - François Hindré
- Univ AngersUniv Nantes, Inserm, CNRS, CRCINA (Centre de Recherche en Cancérologie et Immunologie Nantes—Angers)—UMR 1232, ERL 6001, Nantes, France
- Univ AngersPRIMEX (Plateforme de Radiobiologie et d'Imagerie EXperimentale), Angers, France
| | - Emmanuel Garcion
- Univ AngersUniv Nantes, Inserm, CNRS, CRCINA (Centre de Recherche en Cancérologie et Immunologie Nantes—Angers)—UMR 1232, ERL 6001, Nantes, France
- Univ AngersPRIMEX (Plateforme de Radiobiologie et d'Imagerie EXperimentale), Angers, France
| | - Michel Chérel
- Univ AngersUniv Nantes, Inserm, CNRS, CRCINA (Centre de Recherche en Cancérologie et Immunologie Nantes—Angers)—UMR 1232, ERL 6001, Nantes, France
- ICO (Institut de Cancérologie de l'Ouest)Comprehensive Cancer Center René Gauducheau, Saint-Herblain, France
| | - Nicolas Noiret
- Univ RennesCNRS, ISCR (Institut des Sciences Chimiques de Rennes)—UMR 6226, Rennes, France
- ENSCR (Ecole Nationale Supérieure de Chimie de Rennes)Rennes, France
| | - Etienne Garin
- Comprehensive Cancer Center Eugène MarquisRennes, France
- Univ RennesInra, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer)—UMR_A 1341, UMR_S 1241, Rennes, France
| | - F. F. Russ Knapp
- EmeritusMedical Radioisotopes Program, ORNL (Oak Ridge National Laboratory), Oak Ridge, TN, United States
| |
Collapse
|
13
|
Ersöz OA, Spink R, Griswold JR, Yurt F, Mirzadeh S. Measurement of neutron capture cross section of 187W for production of 188W. Appl Radiat Isot 2019; 148:191-196. [PMID: 30978655 DOI: 10.1016/j.apradiso.2019.03.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 10/27/2022]
Abstract
Tungsten-188 (t1/2 = 69.4 d) is routinely produced by double neutron capture using highly enriched 186W target, 186W(n,γ)187W(n,γ)188W reaction, at the ORNL 85 MWt High Flux Isotope Reactor. While the thermal neutron cross section for the first reaction, 186W(n,γ)187W, is well known, the single reported 64 b cross-section for the second reaction, 187W(n,γ)188W, cannot be validated by experimental results that yield lower than expected activities of 188W. In this study, we report a new value for the thermal neutron capture cross section of 187W. After confirming the neutron capture cross section of 186W (σ0 = 37.8 ± 1.8 b for thermal and I0 = 476 ± 25 b for resonance integrals with σ0/I0 = 12.6 ± 0.4) in two short irradiations, longer irradiations (1-10 d) were performed to obtain a value of 6.5 ± 0.8 b for the σ0 of 187W, which is lower than the adopted value by a factor of 10. Due to the short half-life of 187W (t1/2 = 23.7 d), the σ0 for 187W was obtained empirically by comparing the 188W experimental yields with the theoretical yields generated by code IsoChain and varying the 187W cross section while keeping all other parameters constant.
Collapse
Affiliation(s)
- Onur Alp Ersöz
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100, Izmir, Turkey; Department of Nuclear Engineering & Radiological Sciences, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Robyn Spink
- Department of Nuclear Engineering & Radiological Sciences, University of Michigan, Ann Arbor, MI, 48109, United States; Isotopes and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, United States
| | - Justin R Griswold
- Isotopes and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, United States.
| | - Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100, Izmir, Turkey.
| | - Saed Mirzadeh
- Isotopes and Fuel Cycle Technology Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, United States
| |
Collapse
|
14
|
Abstract
Nuclear medicine is composed of two complementary areas, imaging and therapy. Positron emission tomography (PET) and single-photon imaging, including single-photon emission computed tomography (SPECT), comprise the imaging component of nuclear medicine. These areas are distinct in that they exploit different nuclear decay processes and also different imaging technologies. In PET, images are created from the 511 keV photons produced when the positron emitted by a radionuclide encounters an electron and is annihilated. In contrast, in single-photon imaging, images are created from the γ rays (and occasionally X-rays) directly emitted by the nucleus. Therapeutic nuclear medicine uses particulate radiation such as Auger or conversion electrons or β- or α particles. All three of these technologies are linked by the requirement that the radionuclide must be attached to a suitable vector that can deliver it to its target. It is imperative that the radionuclide remain attached to the vector before it is delivered to its target as well as after it reaches its target or else the resulting image (or therapeutic outcome) will not reflect the biological process of interest. Radiochemistry is at the core of this process, and radiometals offer radiopharmaceutical chemists a tremendous range of options with which to accomplish these goals. They also offer a wide range of options in terms of radionuclide half-lives and emission properties, providing the ability to carefully match the decay properties with the desired outcome. This Review provides an overview of some of the ways this can be accomplished as well as several historical examples of some of the limitations of earlier metalloradiopharmaceuticals and the ways that new technologies, primarily related to radionuclide production, have provided solutions to these problems.
Collapse
Affiliation(s)
- Eszter Boros
- Department of Chemistry , Stony Brook University , Stony Brook , New York 11794 , United States
| | - Alan B Packard
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology , Boston Children's Hospital , Boston , Massachusetts 02115 , United States.,Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
15
|
Guo R, Xi Y, Zhang M, Miao Y, Zhang M, Li B. Human sodium iodide transporter gene-mediated imaging and therapy of mouse glioma, comparison between 188Re and 131I. Oncol Lett 2018; 15:3911-3917. [PMID: 29467904 DOI: 10.3892/ol.2018.7752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 11/16/2017] [Indexed: 01/26/2023] Open
Abstract
Novel treatment options are urgently required for patients with glioma who are not effectively treated through standard therapy. Human sodium iodide symporter (hNIS) is a molecular target of certain tumors types. Compared with 131I, 188Re possesses a higher energy and shorter half-life; therefore, the effects of 188Re and 131I were compared in hNIS-mediated gene imaging and therapy in the present study. Recombinant human brain glioma cell line U87 was transfected with a recombinant lentiviral vector containing hNIS (U87-hNIS). U87-0 cell line transfected with blank lentivirus was prepared as a control. In vitro, the 188Re and 131I uptake of U87-hNIS cells were 21.3-times and 25.9-times that of the control groups, however the excretion rate of the two nuclides was very rapid, and the half-life was only ~4 min. Sodium perchlorate inhibited hNIS-mediated 188Re and 131I uptake to levels observed in the control groups. 188Re and 131I were able to kill U87-hNIS cells selectively, with a survival of only 21.6 and 36.2%, respectively. U87-hNIS nude mice appeared to accumulate 188Re, with a ratio of radioactivity counts between tumor and non-tumor sites of ~13.5 compared with 10.3 of 131I 1 h after radionuclide injection. In contrast with in vitro studies, U87-hNIS cells demonstrated a notable increase in 188Re retention in vivo, even 24 h after 188Re injection. U87-hNIS cells also exhibited increased 131I retention in vivo; however, as the time increased, 131I was rapidly released with the tumor no longer able to be imaged 24 h after 131I injection. Following treatment, U87-hNIS tumors experienced a volume reduction of 24.1%, whereas U87-0 cells demonstrated an increase of 28.8%. 188Re and 131I were revealed to be effective at decreasing tumor volume compared with the control. However, 188Re was significantly more potent compared with 131I (P<0.01). The present study indicated that the U87-hNIS cell line is sufficient to induce specific 188Re and 131I uptake, which may kill cells in vitro and in vivo. 188Re exhibited an increased retention time in vivo compared with 131I, which facilitates the imaging and therapy of U87-hNIS tumors.
Collapse
Affiliation(s)
- Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Yun Xi
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| |
Collapse
|
16
|
Lacoeuille F, Arlicot N, Faivre-Chauvet A. Targeted alpha and beta radiotherapy: An overview of radiopharmaceutical and clinical aspects. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2018. [DOI: 10.1016/j.mednuc.2017.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Pascali G, Matesic L, Zhang B, King AT, Robinson AJ, Ung AT, Fraser BH. Sulfur - fluorine bond in PET radiochemistry. EJNMMI Radiopharm Chem 2017; 2:9. [PMID: 29503850 PMCID: PMC5824700 DOI: 10.1186/s41181-017-0028-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/14/2017] [Indexed: 12/02/2022] Open
Abstract
The importance of the sulfur-fluorine bond is starting to increase in modern medicinal chemistry literature. This is due to a better understanding of the stability and reactivity of this moiety depending on the various oxidation states of sulfur. Furthermore, several commercial reagents used for mild and selective fluorination of organic molecules are based on the known reactivity of S-F groups. In this review, we will show how these examples are translating into the 18F field, both for use as stable tags in finished radiopharmaceuticals and as mildly reactive fluoride-relay intermediates. Finally, we also discuss current opportunities where examples of non-radioactive S-F applications/chemistry may be translated into future 18F radiochemistry applications.
Collapse
Affiliation(s)
- Giancarlo Pascali
- 1Australian Nuclear Science and Technology Organisation, New South Wales, Australia.,2Brain and Mind Centre, University of Sydney, New South Wales, Australia
| | - Lidia Matesic
- 1Australian Nuclear Science and Technology Organisation, New South Wales, Australia
| | - Bo Zhang
- 1Australian Nuclear Science and Technology Organisation, New South Wales, Australia.,3School of Chemistry, Monash University, Victoria, Australia
| | - Andrew T King
- 1Australian Nuclear Science and Technology Organisation, New South Wales, Australia.,4School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW Australia
| | | | - Alison T Ung
- 4School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW Australia
| | - Benjamin H Fraser
- 1Australian Nuclear Science and Technology Organisation, New South Wales, Australia
| |
Collapse
|
18
|
Khoshnevisan A, Chuamsaamarkkee K, Boudjemeline M, Jackson A, Smith GE, Gee AD, Fruhwirth GO, Blower PJ. 18F-Fluorosulfate for PET Imaging of the Sodium-Iodide Symporter: Synthesis and Biologic Evaluation In Vitro and In Vivo. J Nucl Med 2017; 58:156-161. [PMID: 27539841 PMCID: PMC6233868 DOI: 10.2967/jnumed.116.177519] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
Anion transport by the human sodium-iodide symporter (hNIS) is an established target for molecular imaging and radionuclide therapy. Current radiotracers for PET of hNIS expression are limited to 124I- and 18F-BF4- We sought new 18F-labeled hNIS substrates offering higher specific activity, higher affinity, and simpler radiochemical synthesis than 18F-BF4- METHODS: The ability of a range of anions, some containing fluorine, to block 99mTcO4- uptake in hNIS-expressing cells was measured. SO3F- emerged as a promising candidate. 18F-SO3F- was synthesized by reaction of 18F- with SO3-pyridine complex in MeCN and purified using alumina and quaternary methyl ammonium solid-phase extraction cartridges. Chemical and radiochemical purity and serum stability were determined by radiochromatography. Radiotracer uptake and efflux in hNIS-transduced HCT116-C19 cells and the hNIS-negative parent cell line were evaluated in vitro in the presence and absence of a known competitive inhibitor (NaClO4). PET/CT imaging and ex vivo biodistribution measurement were conducted on BALB/c mice, with and without NaClO4 inhibition. RESULTS Fluorosulfate was identified as a potent inhibitor of 99mTcO4- uptake via hNIS in vitro (half-maximal inhibitory concentration, 0.55-0.56 μM (in comparison with 0.29-4.5 μM for BF4-, 0.07 μM for TcO4-, and 2.7-4.7 μM for I-). Radiolabeling to produce 18F-SO3F- was simple and afforded high radiochemical purity suitable for biologic evaluation (radiochemical purity > 95%, decay-corrected radiochemical yield = 31.6%, specific activity ≥ 48.5 GBq/μmol). Specific, blockable hNIS-mediated uptake in HCT116-C19 cells was observed in vitro, and PET/CT imaging of normal mice showed uptake in thyroid, salivary glands (percentage injected dose/g at 30 min, 563 ± 140 and 32 ± 9, respectively), and stomach (percentage injected dose/g at 90 min, 68 ± 21). CONCLUSION Fluorosulfate is a high-affinity hNIS substrate. 18F-SO3F- is easily synthesized in high yield and very high specific activity and is a promising candidate for preclinical and clinical PET imaging of hNIS expression and thyroid-related disease; it is the first example of in vivo PET imaging with a tracer containing an S-18F bond.
Collapse
Affiliation(s)
- Alex Khoshnevisan
- Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, King's College London, London, United Kingdom; and
| | - Krisanat Chuamsaamarkkee
- Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, King's College London, London, United Kingdom; and
| | - Mehdi Boudjemeline
- Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, King's College London, London, United Kingdom; and
| | | | | | - Antony D Gee
- Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, King's College London, London, United Kingdom; and
| | - Gilbert O Fruhwirth
- Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, King's College London, London, United Kingdom; and
| | - Philip J Blower
- Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, King's College London, London, United Kingdom; and
| |
Collapse
|
19
|
Khoshnevisan A, Jauregui-Osoro M, Shaw K, Torres JB, Young JD, Ramakrishnan NK, Jackson A, Smith GE, Gee AD, Blower PJ. [(18)F]tetrafluoroborate as a PET tracer for the sodium/iodide symporter: the importance of specific activity. EJNMMI Res 2016; 6:34. [PMID: 27103614 PMCID: PMC4840125 DOI: 10.1186/s13550-016-0188-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/11/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND [(18)F]BF4 (-), the first (18)F-labelled PET imaging agent for the sodium/iodide symporter (NIS), was produced by isotopic exchange yielding a product with limited specific activity (SA, ca. 1 GBq/μmol) posing a risk of sub-optimal target-to-background ratios (TBR) in PET images due to saturation of NIS in vivo. We sought to quantify this risk and to develop a method of production of [(18)F]BF4 (-) with higher SA. METHODS A new radiosynthesis of [(18)F]BF4 (-) was developed, involving reaction of [(18)F]F(-) with boron trifluoride diethyl etherate under anhydrous conditions, guided by (11)B and (19)F NMR studies of equilibria involving BF4 (-) and BF3. The SA of the product was determined by ion chromatography. The IC50 of [(19)F]BF4 (-) as an inhibitor of [(18)F]BF4 (-) uptake was determined in vitro using HCT116-C19 human colon cancer cells expressing the human form of NIS (hNIS). The influence of [(19)F]BF4 (-) dose on biodistribution in vivo was evaluated in normal mice by nanoPET imaging and ex vivo tissue counting. RESULTS An IC50 of 4.8 μΜ was found in vitro indicating a significant risk of in vivo NIS saturation at SA achieved by the isotopic exchange labelling method. In vivo thyroid and salivary gland uptake decreased significantly with [(19)F]BF4 (-) doses above ca. 10 μg/kg. The new radiosynthesis gave high radiochemical purity (>99 %) and moderate yield (15 %) and improved SA (>5 GBq/μmol) from a starting activity of only 1.5 GBq. CONCLUSIONS [(18)F]BF4 (-) produced at previously reported levels of SA (1 GBq/μmol) can lead to reduced uptake in NIS-expressing tissues in mice. This is much less likely in humans. The synthetic approach described provides an alternative for production of [(18)F]BF4 (-) at higher SA with sufficient yield and without need for unusually high starting activity of [(18)F]fluoride, removing the risk of NIS saturation in vivo even in mice. TRIAL REGISTRATION ISRCTN75827286 .
Collapse
Affiliation(s)
- Alex Khoshnevisan
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Maite Jauregui-Osoro
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Karen Shaw
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Julia Baguña Torres
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Jennifer D Young
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Nisha K Ramakrishnan
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Alex Jackson
- The Grove Centre, GE Healthcare, White Lion Road, Amersham, UK
| | - Gareth E Smith
- The Grove Centre, GE Healthcare, White Lion Road, Amersham, UK
| | - Antony D Gee
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Philip J Blower
- Division of Imaging Sciences and Biomedical Engineering, King's College London, 4th Floor Lambeth Wing, St. Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
20
|
Use of rhenium-188 for in vivo imaging and treatment of human cervical cancer cells transfected with lentivirus expressing sodium iodide symporter. Oncol Rep 2016; 36:2289-97. [PMID: 27573304 DOI: 10.3892/or.2016.5034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 08/16/2016] [Indexed: 11/05/2022] Open
Abstract
Although survival rates for cervical cancer have improved, they need further improvement in patients with distant metastases. The sodium iodine symporter (NIS) gene has often been used in cancer therapy and imaging. We examined the therapeutic effects of rhenium-188 (188Re) in a cervical cancer xenograft model expressing the NIS gene under the control of the tumor-specific human telomerase reverse transcriptase (hTERT) promoter. We constructed two recombinant lentiviral vectors expressing enhanced green fluorescent protein (eGFP) or the NIS gene driven by the hTERT promoter. To determine the tumor-specific transcriptional activity of the hTERT promoter, the eGFP-expressing vector was stably transfected into tumor cells and normal cells. A cervical cancer HeLa cell line stably expressing NIS (HeLa-TERTNIS) was created and examined in a similar way. HeLa and HeLa-TERTNIS tumor xenografts were transplanted in nude mice, and in vivo 188Re distribution was measured using micro-SPECT/CT imaging. The therapeutic effects of 188Re were assessed over 21 days on the basis of tumor volume and the immunohistochemical findings of excised tumors. eGFP expression controlled by the hTERT promoter was substantially higher in the tumor cells than normal cells. Quantitative PCR and western blotting confirmed that HeLa-TERTNIS cells expressed high levels of NIS mRNA and protein, respectively. Further, 188Re uptake and accumulation were significantly higher in HeLa-TERTNIS cells and xenografts than HeLa cells and xenografts. In vitro and in vivo, 188Re significantly reduced the survival of HeLa-TERTNIS cells and inhibited the growth of HeLa-TERTNIS xenografts, respectively. Immunohistochemical staining showed that HeLa-TERTNIS xenograft tumors expressed higher levels of NIS and caspase-3 and lower levels of Ki-67 than HeLa xenograft tumors. Our findings indicated that hTERT promoter-driven expression of the NIS gene in HeLa cells led to 188Re uptake and therapeutic effects. Thus, NIS-based gene therapy and imaging using the hTERT promoter and 188Re may be possible.
Collapse
|
21
|
|
22
|
Guo R, Zhang M, Xi Y, Ma Y, Liang S, Shi S, Miao Y, Li B. Theranostic studies of human sodium iodide symporter imaging and therapy using 188Re: a human glioma study in mice. PLoS One 2014; 9:e102011. [PMID: 25000403 PMCID: PMC4084984 DOI: 10.1371/journal.pone.0102011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/13/2014] [Indexed: 11/18/2022] Open
Abstract
Objective To investigate the role of 188Re in human sodium iodide symporter (hNIS) theranostic gene-mediated human glioma imaging and therapy in model mice. Methods The human glioma cell line U87 was transfected with recombinant lentivirus encoding the hNIS gene under the control of cytomegalovirus promoter (U87-hNIS). The uptake and efflux of 188Re were determined after incubating the cells with 188Re. 188Re uptake experiments in the presence of various concentrations of sodium perchlorate were carried out. In vitro cell killing tests with 188Re were performed. U87-hNIS mediated 188Re distribution, imaging and therapy in nude mice were also tested. Results U87-hNIS cell line was successfully established. The uptake of 188Re in U87-hNIS cells increased up to 26-fold compared to control cells, but was released rapidly with a half-life of approximately 4 minutes. Sodium perchlorate reduced hNIS-mediated 188Re uptake to levels of control cell lines. U87-hNIS cells were selectively killed following exposure to 188Re, with a survival of 21.4%, while control cells had a survival of 92.1%. Unlike in vitro studies, U87-hNIS tumor showed a markedly increased 188Re retention even 48 hours after 188Re injection. In the therapy study, there was a significant difference in tumor size between U87-hNIS mice (317±67 mm3) and control mice (861±153 mm3) treated with 188Re for 4 weeks (P<0.01). Conclusion The results indicate that inserting the hNIS gene into U87 cells is sufficient to induce specific 188Re uptake, which has a cell killing effect both in vitro and in vivo. Moreover, our study, based on the function of hNIS as a theranostic gene allowing noninvasive imaging of hNIS expression by 188Re scintigraphy, provides detailed characterization of in vivo vector biodistribution and level, localization, essential prerequisites for precise planning and monitoring of clinical gene therapy that aims to individualize gene therapy concept.
Collapse
Affiliation(s)
- Rui Guo
- Department of Nuclear Medicine, Rui Jin Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - M. Zhang
- Department of Nuclear Medicine, Rui Jin Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yun Xi
- Department of Nuclear Medicine, Rui Jin Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yufei Ma
- Department of Nuclear Medicine, Xin Hua Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - Sheng Liang
- Department of Nuclear Medicine, Xin Hua Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - Shuo Shi
- Department of Nuclear Medicine, Rui Jin Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Rui Jin Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Rui Jin Hospital, School of medicine, Shanghai JiaoTong University, Shanghai, China
- * E-mail:
| |
Collapse
|
23
|
Portulano C, Paroder-Belenitsky M, Carrasco N. The Na+/I- symporter (NIS): mechanism and medical impact. Endocr Rev 2014; 35:106-49. [PMID: 24311738 PMCID: PMC3895864 DOI: 10.1210/er.2012-1036] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 10/11/2013] [Indexed: 12/26/2022]
Abstract
The Na(+)/I(-) symporter (NIS) is the plasma membrane glycoprotein that mediates active I(-) transport in the thyroid and other tissues, such as salivary glands, stomach, lactating breast, and small intestine. In the thyroid, NIS-mediated I(-) uptake plays a key role as the first step in the biosynthesis of the thyroid hormones, of which iodine is an essential constituent. These hormones are crucial for the development of the central nervous system and the lungs in the fetus and the newborn and for intermediary metabolism at all ages. Since the cloning of NIS in 1996, NIS research has become a major field of inquiry, with considerable impact on many basic and translational areas. In this article, we review the most recent findings on NIS, I(-) homeostasis, and related topics and place them in historical context. Among many other issues, we discuss the current outlook on iodide deficiency disorders, the present stage of understanding of the structure/function properties of NIS, information gleaned from the characterization of I(-) transport deficiency-causing NIS mutations, insights derived from the newly reported crystal structures of prokaryotic transporters and 3-dimensional homology modeling, and the novel discovery that NIS transports different substrates with different stoichiometries. A review of NIS regulatory mechanisms is provided, including a newly discovered one involving a K(+) channel that is required for NIS function in the thyroid. We also cover current and potential clinical applications of NIS, such as its central role in the treatment of thyroid cancer, its promising use as a reporter gene in imaging and diagnostic procedures, and the latest studies on NIS gene transfer aimed at extending radioiodide treatment to extrathyroidal cancers, including those involving specially engineered NIS molecules.
Collapse
Affiliation(s)
- Carla Portulano
- Department of Molecular and Cellular Physiology (C.P., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; and Department of Molecular Pharmacology (M.P.-B.), Albert Einstein College of Medicine, Bronx, New York 10469
| | | | | |
Collapse
|
24
|
Knoop K, Schwenk N, Dolp P, Willhauck MJ, Zischek C, Zach C, Hacker M, Göke B, Wagner E, Nelson PJ, Spitzweg C. Stromal targeting of sodium iodide symporter using mesenchymal stem cells allows enhanced imaging and therapy of hepatocellular carcinoma. Hum Gene Ther 2013; 24:306-16. [PMID: 23402366 DOI: 10.1089/hum.2012.104] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The tumor-homing property of mesenchymal stem cells (MSC) has lead to their use as delivery vehicles for therapeutic genes. The application of the sodium iodide symporter (NIS) as therapy gene allows noninvasive imaging of functional transgene expression by (123)I-scintigraphy or PET-imaging, as well as therapeutic application of (131)I or (188)Re. Based on the critical role of the chemokine RANTES (regulated on activation, normal T-cell expressed and presumably secreted)/CCL5 secreted by MSCs in the course of tumor stroma recruitment, use of the RANTES/CCL5 promoter should allow tumor stroma-targeted expression of NIS after MSC-mediated delivery. Using a human hepatocellular cancer (HCC) xenograft mouse model (Huh7), we investigated distribution and tumor recruitment of RANTES-NIS-engineered MSCs after systemic injection by gamma camera imaging. (123)I-scintigraphy revealed active MSC recruitment and CCL5 promoter activation in the tumor stroma of Huh7 xenografts (6.5% ID/g (123)I, biological half-life: 3.7 hr, tumor-absorbed dose: 44.3 mGy/MBq). In comparison, 7% ID/g (188)Re was accumulated in tumors with a biological half-life of 4.1 hr (tumor-absorbed dose: 128.7 mGy/MBq). Administration of a therapeutic dose of (131)I or (188)Re (55.5 MBq) in RANTES-NIS-MSC-treated mice resulted in a significant delay in tumor growth and improved survival without significant differences between (131)I and (188)Re. These data demonstrate successful stromal targeting of NIS in HCC tumors by selective recruitment of NIS-expressing MSCs and by use of the RANTES/CCL5 promoter. The resulting tumor-selective radionuclide accumulation was high enough for a therapeutic effect of (131)I and (188)Re opening the exciting prospect of NIS-mediated radionuclide therapy of metastatic cancer using genetically engineered MSCs as gene delivery vehicles.
Collapse
Affiliation(s)
- Kerstin Knoop
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rhenium-188 production in hospitals, by w-188/re-188 generator, for easy use in radionuclide therapy. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2013; 2013:290750. [PMID: 23653859 PMCID: PMC3638650 DOI: 10.1155/2013/290750] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 03/13/2013] [Indexed: 11/17/2022]
Abstract
Rhenium-188 (Re-188) is a high energy β-emitting radioisotope obtained from the tungsten-188/rhenium-188 (W-188/Re-188) generator, which has shown utility for a variety of therapeutic applications in nuclear medicine, oncology, and interventional radiology/cardiology. Re-188 decay is accompanied by a 155 keV predominant energy γ-emission, which could be detected by γ-cameras, for imaging, biodistribution, or absorbed radiation dose studies. Its attractive physical properties and its potential low cost associated with a long-lived parent make it an interesting option for clinical use. The setup and daily use of W-188/Re-188 generator in hospital nuclear medicine departments are discussed in detail. The clinical efficacy, for several therapeutic applications, of a variety of Re-188-labeled agents is demonstrated. The high energy of the β-emission of Re-188 is particularly well suited for effective penetration in solid tumours. Its total radiation dose delivered to tissues is comparable to other radionuclides used in therapy. Furthermore, radiation safety and shielding requirements are an important subject of matter. In the case of bone metastases treatment, therapeutic ratios are presented in order to describe the efficacy of Re-188 usage.
Collapse
|
26
|
So Y, Lee YJ, Lee WW, Chung JK. Determination of the optimal time for radioiodine therapy in anaplastic thyroid carcinoma using the adenovirus-mediated transfer of sodium iodide symporter gene. Oncol Rep 2013; 29:1666-70. [PMID: 23404348 DOI: 10.3892/or.2013.2277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/28/2012] [Indexed: 11/06/2022] Open
Abstract
Gene therapy using human sodium iodide symporter (hNIS) and radioiodine has been considered promising in a variety of gene therapy trials. However, the optimal timing of radioiodine application following hNIS gene transfer remains unknown. The present study aimed to investigate the serial expression of hNIS following adenovirus-mediated hNIS gene transfer into anaplastic thyroid carcinoma (ARO) to determine the optimal timing of radioiodine application. Recombinant adenovirus encoding the hNIS gene (rAd-hNIS) was generated using a homologous recombination reaction. The iodine uptake of rAd-hNIS‑transfected ARO cells gradually increased until 120 min post‑125I application but the fold increase, reflecting the relative uptake of rAd-hNIS‑transfected compared to non‑transfected ARO cells, reached plateau at 60 min post‑125I application. For the in vivo analysis, rAd-hNIS was injected intratumorally into ARO cell xenografts in the thighs of nude mice (n=12). Two, 3, 4 and 6 days after rAd-hNIS injection, γ‑scintigraphic images were obtained 60 min following injection of 5.5 MBq of 131I intraperitoneally. Treated/non-treated (T/NT) xenograft count ratios were the highest at day 2 post‑rAd-hNIS injection (2.85±0.61), and gradually decreased thereafter (2.54±0.65, 2.31±0.42 and 2.18±0.90 at days 3, 4 and 6 post‑rAd-hNIS injection, respectively). Real‑time polymerase chain reaction (RT-PCR) and immunohistochemical staining demonstrated that hNIS expression was the highest at day 2 following rAd-hNIS injection. In conclusion, the optimal timing for radioiodine administration is day 2 after adenovirus-mediated hNIS gene transfer into anaplastic thyroid carcinoma.
Collapse
Affiliation(s)
- Young So
- Department of Nuclear Medicine, Konkuk University School of Medicine, Chungju, Republic of Korea
| | | | | | | |
Collapse
|
27
|
Kim KI, Lee YJ, Lee TS, Song I, Cheon GJ, Lim SM, Chung JK, Kang JH. In vitro radionuclide therapy and in vivo scintigraphic imaging of alpha-fetoprotein-producing hepatocellular carcinoma by targeted sodium iodide symporter gene expression. Nucl Med Mol Imaging 2012; 47:1-8. [PMID: 24895502 DOI: 10.1007/s13139-012-0166-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/10/2012] [Accepted: 08/20/2012] [Indexed: 12/23/2022] Open
Abstract
PURPOSE This study aimed to develop a gene expression targeting method for specific imaging and therapy of alpha-fetoprotein (AFP)-producing hepatocellular carcinoma (HCC) cells, using an adenovirus vector containing the human sodium/iodide symporter (hNIS) gene driven by an AFP enhancer/promoter. METHODS The recombinant adenovirus vector, AdAFPhNIS (containing the hNIS gene driven by human AFP enhancer/promoter) was prepared. After in vitro infection by the adenovirus, hNIS gene expression in AFP-producing cells and in AFP-nonproducing cells was investigated using (125)I uptake assay and semi-quantitative reverse transcription polymerase chain reaction (RT-PCR). The killing effect of (131)I on AdAFPhNIS-infected HCC cells was studied using an in vitro clonogenic assay. In addition, tumor-bearing mice were intravenously injected with the adenovirus, and scintigraphic images were obtained. RESULTS The expression of hNIS was efficiently demonstrated by (125)I uptake assay in AFP-producing cells, but not in AFP-nonproducing cells. AFP-producing HCC-targeted gene expression was confirmed at the mRNA level. Furthermore, in vitro clonogenic assay showed that hNIS gene expression induced by AdAFPhNIS infection in AFP-producing cells caused more sensitivity to (131)I than that in AFP-nonproducing cells. Injected intravenously in HuH-7 tumor xenografts mice by adenovirus, the functional hNIS gene expression was confirmed in tumor by in vivo scintigraphic imaging. CONCLUSIONS An AFP-producing HCC was targeted with an adenovirus vector containing the hNIS gene using the AFP enhancer/promoter in vitro and in vivo. These findings demonstrate that AFP-producing HCC-specific molecular imaging and radionuclide gene therapy are feasible using this recombinant adenovirus vector system.
Collapse
Affiliation(s)
- Kwang Il Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-gil, Gongneung-dong, Nowon-gu, Seoul 139-706 Korea
| | - Yong Jin Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-gil, Gongneung-dong, Nowon-gu, Seoul 139-706 Korea
| | - Tae Sup Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-gil, Gongneung-dong, Nowon-gu, Seoul 139-706 Korea
| | - Inho Song
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-gil, Gongneung-dong, Nowon-gu, Seoul 139-706 Korea
| | - Gi Jeong Cheon
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-gil, Gongneung-dong, Nowon-gu, Seoul 139-706 Korea ; Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Sang Moo Lim
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, 28, Yongon-dong, Jongno-gu, Seoul 110-744 Korea
| | - Joo Hyun Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-gil, Gongneung-dong, Nowon-gu, Seoul 139-706 Korea
| |
Collapse
|
28
|
Mu D, Huang R, Li S, Ma X, Lou C, Kuang A. Combining transfer of TTF-1 and Pax-8 gene: a potential strategy to promote radioiodine therapy of thyroid carcinoma. Cancer Gene Ther 2012; 19:402-11. [PMID: 22498723 DOI: 10.1038/cgt.2012.13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cotransfer of thyroid-specific transcription factor (TTF)-1 and Pax-8 gene to tumor cells, resulting in the re-expression of iodide metabolism-associated proteins, such as sodium iodide symporter (NIS), thyroglobulin (Tg), thyroperoxidase (TPO), offers the possibility of radioiodine therapy to non-iodide-concentrating tumor because the expression of iodide metabolism-associated proteins in thyroid are mediated by the thyroid transcription factor TTF-1 and Pax-8. The human TTF-1 and Pax-8 gene were transducted into the human thyroid carcinoma (K1 and F133) cells by the recombinant adenovirus, AdTTF-1 and AdPax-8. Re-expression of NIS mRNA and protein, but not TPO and Tg mRNA and protein, was detected in AdTTF-1-infected F133 cells, following with increasing radioiodine uptake (6.1-7.4 times), scarcely iodide organification and rapid iodide efflux (t(1/2) ≈ 8-min in vitro, t(1/2) ≈ 4.7-h in vivo). On contrast, all of the re-expression of NIS, TPO and Tg mRNA and proteins were detected in F133 cells coinfected with AdTTF-1 and AdPax-8. AdTTF-1- and AdPax-8-coinfected K1 and F133 cells could effectively accumulate radioiodine (6.6-7.5 times) and obviously retarded radioiodine retention (t(1/2) ≈ 25-30-min in vitro, t(1/2) ≈ 12-h in vivo) (P<0.05). Accordingly, the effect of radioiodine therapy of TTF-1 and Pax-8 cotransducted K1 and F133 cells (21-25% survival rate in vitro) was better than that of TTF-1-transducted cells (40% survival rate in vitro) (P<0.05). These results indicate that single TTF-1 gene transfer may have limited efficacy of radioiodine therapy because of rapid radioiodine efflux. The cotransduction of TTF-1 and Pax-8 gene, with resulting NIS-mediated radioiodine accumulation and TPO and Tg-mediated radioiodine organification and intracellular retention, may lead to effective radioiodine therapy of thyroid carcinoma.
Collapse
Affiliation(s)
- D Mu
- Department of Nuclear Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
29
|
Mu D, Huang R, Ma X, Li S, Kuang A. Radioiodine therapy of thyroid carcinoma following Pax-8 gene transfer. Gene Ther 2011; 19:435-42. [PMID: 21833035 DOI: 10.1038/gt.2011.110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The thyroid transcription factor Pax-8 could bind with the promoter/enhancer of thyroid-specific genes such as thyroglobulin (Tg), thyroperoxidase (TPO) and sodium iodide symporter (NIS), and regulate the expression of these proteins in thyrocyte. Promoting iodide accumulation in tumor cells by re-expression of Pax-8 provides a possible strategy for radioiodine therapy of tumor. Therefore, we investigated the effect of Pax-8 gene transfer on radioiodine therapy of thyroid carcinoma. The human Pax-8 gene was transfected into the human thyroid carcinoma (K1 and F133) cells by the recombinant adenovirus vector. Although the NIS mRNA was not detected, the expression of mRNA and proteins of Tg and TPO in AdPax-8-infected F133 cells were activated by Pax-8. Iodide uptake in thyroid carcinoma cells was reactivated by Pax-8 (increasing 3.3-fold in K1 cells and 5.7-fold in F133 cells). Moreover, Pax-8 promoted iodide organification and the retention time of iodine in Pax-8-expressing cells apparently prolonged in vitro and in vivo (P<0.05). Pax-8-expressing thyroid carcinoma cells were selectively killed by radioiodine. The AdPax-8-infected tumors in vivo clearly visualized in scanning images at 12 h after administration of radioiodine. These results indicate that Pax-8 can promote iodide uptake, and specifically prolong the retention time of iodide in thyroid cancer in vitro and in vivo by promoting the expression of TPO and Tg proteins. Pax-8 gene transfection may lead to effective radioiodine therapy of tumor.
Collapse
Affiliation(s)
- D Mu
- Department of Nuclear Medicine, National Key Discipline of Medical Imaging and Nuclear medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | | | | |
Collapse
|
30
|
Lacoeuille F, Hindré F, Venier-Julienne MC, Sergent M, Bouchet F, Jouaneton S, Denizot B, Askienazy S, Benoit JP, Couturier OF, Le Jeune JJ. A starch-based microparticulate system dedicated to diagnostic and therapeutic nuclear medicine applications. Biomaterials 2011; 32:7999-8009. [PMID: 21788070 DOI: 10.1016/j.biomaterials.2011.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 07/04/2011] [Indexed: 01/18/2023]
Abstract
The aim of this work was to develop a new microparticulate system able to form a complex with radionuclides with a high yield of purity for diagnostic or therapeutic applications. Owing to its properties potato starch was chosen as starting material and modified by oxidization and coupling of a ligand (polyamine) enabling modified starch to chelate radionuclides. The choice of suitable experiments was based on a combination of a Rechtschaffner experimental design and a surface response design to determine the influence of experimental parameters and to optimize the final product. Starch-based microparticle formulations from the experimental plans were compared and characterized through particle size analysis, scanning electron microscopy, elemental analysis and, for the most promising formulations, by in vitro labeling stability studies and determination of free polyamine content or in vivo imaging studies. The mechanism of starch-based microparticle degradation was identified by means of size measurements. The results of the Rechtschaffner design showed the positive qualitative effect of the temperature and the duration of coupling reaction whereas surface response analysis clearly showed that, by increasing the oxidization level and starch concentration, the nitrogen content in the final product is increased. In vitro and in vivo characterization led to identification of the best formulation. With a size around 30 μm, high radiochemical purity (over 95%) and a high signal-to-noise ratio (over 600), the new starch-based microparticulate system could be prepared as ready-to-use kits and sterilized without modification of its characteristics, and thus meet the requirement for in vivo diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- F Lacoeuille
- LUNAM Université, Université d'Angers, Inserm U646, Centre Hospitalier Universitaire d'Angers, 4 rue Larrey, F 49100 Angers, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Leggett RW. A Physiological Systems Model for Iodine for Use in Radiation Protection. Radiat Res 2010; 174:496-516. [DOI: 10.1667/rr2243.1] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
32
|
Hingorani M, Spitzweg C, Vassaux G, Newbold K, Melcher A, Pandha H, Vile R, Harrington K. The biology of the sodium iodide symporter and its potential for targeted gene delivery. Curr Cancer Drug Targets 2010; 10:242-67. [PMID: 20201784 DOI: 10.2174/156800910791054194] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 02/16/2010] [Indexed: 12/12/2022]
Abstract
The sodium iodide symporter (NIS) is responsible for thyroidal, salivary, gastric, intestinal and mammary iodide uptake. It was first cloned from the rat in 1996 and shortly thereafter from human and mouse tissue. In the intervening years, we have learned a great deal about the biology of NIS. Detailed knowledge of its genomic structure, transcriptional and post-transcriptional regulation and pharmacological modulation has underpinned the selection of NIS as an exciting approach for targeted gene delivery. A number of in vitro and in vivo studies have demonstrated the potential of using NIS gene therapy as a means of delivering highly conformal radiation doses selectively to tumours. This strategy is particularly attractive because it can be used with both diagnostic (99mTc, 125I, 124I)) and therapeutic (131I, 186Re, 188Re, 211At) radioisotopes and it lends itself to incorporation with standard treatment modalities, such as radiotherapy or chemoradiotherapy. In this article, we review the biology of NIS and discuss its development for gene therapy.
Collapse
Affiliation(s)
- Mohan Hingorani
- The Institute of Cancer Research, 237 Fulham Road, London SW36JB, UK
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Klutz K, Russ V, Willhauck MJ, Wunderlich N, Zach C, Gildehaus FJ, Göke B, Wagner E, Ogris M, Spitzweg C. Targeted radioiodine therapy of neuroblastoma tumors following systemic nonviral delivery of the sodium iodide symporter gene. Clin Cancer Res 2009; 15:6079-86. [PMID: 19789324 DOI: 10.1158/1078-0432.ccr-09-0851] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We recently reported the significant therapeutic efficacy of radioiodine therapy in various tumor mouse models following transcriptionally targeted sodium iodide symporter (NIS) gene transfer. These studies showed the high potential of NIS as a novel diagnostic and therapeutic gene for the treatment of extrathyroidal tumors. As a next crucial step towards clinical application of NIS-mediated radionuclide therapy we aim at systemic delivery of the NIS gene to target extrathyroidal tumors even in the metastatic stage. EXPERIMENTAL DESIGN In the current study, we used synthetic polymeric vectors based on pseudodendritic oligoamines with high intrinsic tumor affinity (G2-HD-OEI) to target a NIS-expressing plasmid (CMV-NIS-pcDNA3) to neuroblastoma (Neuro2A) cells. RESULTS Incubation with NIS-containing polyplexes (G2-HD-OEI/NIS) resulted in a 51-fold increase in perchlorate-sensitive iodide uptake activity in Neuro2A cells in vitro. Through (123)I-scintigraphy and ex vivo gamma counting Neuro2A tumors in syngeneic A/J mice were shown to accumulate 8% to 13% ID/g (123)I with a biological half-life of 13 hours, resulting in a tumor-absorbed dose of 247 mGy/MBq (131)I after i.v. application of G2-HD-OEI/NIS. Nontarget organs, including liver, lung, kidneys, and spleen revealed no significant iodide uptake. Moreover, two cycles of systemic NIS gene transfer followed by (131)I application (55.5 MBq) resulted in a significant delay in tumor growth associated with markedly improved survival. CONCLUSIONS In conclusion, our data clearly show the high potential of novel pseudodendritic polymers for tumor-specific NIS gene delivery after systemic application, opening the prospect of targeted NIS-mediated radionuclide therapy of nonthyroidal tumors even in metastatic disease.
Collapse
Affiliation(s)
- Kathrin Klutz
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Torres Martin de Rosales R, Årstad E, Blower PJ. Nuclear imaging of molecular processes in cancer. Target Oncol 2009; 4:183-97. [DOI: 10.1007/s11523-009-0120-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Accepted: 09/09/2009] [Indexed: 12/25/2022]
|
35
|
Wild-type p53 enhances the cytotoxic effect of radionuclide gene therapy using sodium iodide symporter in a murine anaplastic thyroid cancer model. Eur J Nucl Med Mol Imaging 2009; 37:235-41. [DOI: 10.1007/s00259-009-1251-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 07/31/2009] [Indexed: 10/20/2022]
|
36
|
Riese CGU, Seitz S, Schipper ML, Behr TM. Effective treatment of pancreatic neuroendocrine tumours transfected with the sodium iodide symporter gene by 186Re-perrhenate in mice. Eur J Nucl Med Mol Imaging 2009; 36:1767-73. [DOI: 10.1007/s00259-009-1153-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Accepted: 04/17/2009] [Indexed: 10/20/2022]
|
37
|
Jin YN, Chung HK, Kang JH, Lee YJ, Kimm KI, Kim YJ, Kim S, Chung JK. Radioiodine gene therapy of hepatocellular carcinoma targeted human alpha fetoprotein. Cancer Biother Radiopharm 2009; 23:551-60. [PMID: 18986218 DOI: 10.1089/cbr.2008.0467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION We conducted a molecular imaging and gene therapy method in alpha-fetoprotein (AFP)-producing hepatocellular carcinoma (HCC) by tumor-specific expression of the human sodium/iodide symporter (hNIS) using an AFP promoter. METHODS The tumor-specific expression of hNIS gene by the AFP enhancer/promoter was constructed as pcDNA3-AFP/hNIS. The pcDNA3-AFP/hNIS was stably transfected to human HCC (Huh-7/AN) and rat glioma cells (C6/AN). Functional hNIS expression was confirmed by radioiodine uptake. The mRNA and protein-expression level of hNIS were measured. Biodistribution of 131I was evaluated, and scintigraphic images of 99mTc were obtained in xenografted mice. A clonogenic assay was performed by 131I. And, the in vivo therapeutic effect of 131I was evaluated in xenografted mice. RESULTS In Huh-7/AN cells, iodine was highly accumulated and completely blocked by perchlorate. The protein and mRNA expression levels were correlated with iodine uptake. Radioiodine uptake in Huh-7/AN tumors was higher than those of control tumors and clearly visualized. The survival rate was significantly decreased in Huh-7/AN cells by 131I. Moreover, a growth of Huh-7/AN tumors was inhibited by 131I in mice. CONCLUSIONS AFP-producing hepatoma can be targeted and treated with radionuclides and hNIS, using AFP enhancer/promoter. This targeted hNIS gene therapy and molecular imaging have the potential to be used in the management of AFP-producing HCC.
Collapse
Affiliation(s)
- Yong Nan Jin
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Assessment of gene function following the completion of human genome sequencing may be done using radionuclide imaging procedures. These procedures are needed for the evaluation of genetically manipulated animals or newly designed biomolecules which require a thorough understanding of physiology, biochemistry and pharmacology. The experimental approaches will involve many new technologies, including in-vivo imaging with SPECT and PET. Nuclear medicine procedures may be applied for the determination of gene function and regulation using established and new tracers or using in-vivo reporter genes, such as genes encoding enzymes, receptors, antigens or transporters. Visualization of in-vivo reporter gene expression can be done using radiolabeled substrates, antibodies or ligands. Combinations of specific promoters and in-vivo reporter genes may deliver information about the regulation of the corresponding genes. Furthermore, protein-protein interactions and the activation of signal transduction pathways may be visualized noninvasively. The role of radiolabeled antisense molecules for the analysis of mRNA content has to be investigated. However, possible applications are therapeutic interventions using triplex oligonucleotides with therapeutic isotopes, which can be brought near to specific DNA sequences to induce DNA strand breaks at selected loci. After the identification of new genes, functional information is required to investigate the role of these genes in living organisms. This can be done by analysis of gene expression, protein-protein interaction or the biodistribution of new molecules and may result in new diagnostic and therapeutic procedures, which include visualization of and interference with gene transcription, and the development of new biomolecules to be used for diagnosis and treatment. Furthermore, the characterization of tumor cell-specific properties allows the design of new treatment modalities, such as gene therapy, which circumvent resistance mechanisms towards conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Uwe Haberkorn
- Department of Nuclear Medicine, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany.
| |
Collapse
|
39
|
Carvalho DP, Ferreira ACF. The importance of sodium/iodide symporter (NIS) for thyroid cancer management. ACTA ACUST UNITED AC 2008; 51:672-82. [PMID: 17891230 DOI: 10.1590/s0004-27302007000500004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 02/17/2007] [Indexed: 01/02/2023]
Abstract
The thyroid gland has the ability to uptake and concentrate iodide, which is a fundamental step in thyroid hormone biosynthesis. Radioiodine has been used as a diagnostic and therapeutic tool for several years. However, the studies related to the mechanisms of iodide transport were only possible after the cloning of the gene that encodes the sodium/iodide symporter (NIS). The studies about the regulation of NIS expression and the possibility of gene therapy with the aim of transferring NIS gene to cells that normally do not express the symporter have also become possible. In the majority of hypofunctioning thyroid nodules, both benign and malignant, NIS gene expression is maintained, but NIS protein is retained in the intracellular compartment. The expression of NIS in non-thyroid tumoral cells in vivo has been possible through the transfer of NIS gene under the control of tissue-specific promoters. Apart from its therapeutic use, NIS has also been used for the localization of metastases by scintigraphy or PET-scan with 124I. In conclusion, NIS gene cloning led to an important development in the field of thyroid pathophysiology, and has also been fundamental to extend the use of radioiodine for the management of non-thyroid tumors.
Collapse
Affiliation(s)
- Denise P Carvalho
- Laboratório de Fisiologia Endócrina, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil.
| | | |
Collapse
|
40
|
Haberkorn U. Modulation of tracer accumulation in malignant tumors: gene expression, gene transfer, and phage display. Curr Top Dev Biol 2008; 70:145-69. [PMID: 16338341 DOI: 10.1016/s0070-2153(05)70007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Assessment of gene function following the completion of human genome sequencing may be done using radionuclide imaging procedures. These procedures are needed for the evaluation of genetically manipulated animals or new designed biomolecules which requires a thorough understanding of physiology, biochemistry and pharmacology. The experimental approaches will involve many new technologies including in vivo imaging with SPECT and PET. Nuclear medicine procedures may be applied for the determination of gene function and regulation using established and new tracers or using in vivo reporter genes such as genes encoding enzymes, receptors, antigens or transporters. Visualization of in vivo reporter gene expression can be done using radiolabeled substrates, antibodies or ligands. Combinations of specific promoters and in vivo reporter genes may deliver information about the regulation of the corresponding genes. Furthermore, protein-protein interactions and activation of signal transduction pathways may be visualized non-invasively. The role of radiolabeled antisense molecules for the analysis of mRNA content has to be investigated. However, possible applications are therapeutic intervention using triplex oligonucleotides with therapeutic isotopes which can be brought near to specific DNA sequences to induce DNA strand breaks at selected loci. Imaging of labeled siRNA's makes sense if these are used for therapeutic purposes in order to assess the delivery of these new drugs to their target tissue. Finally, new biomolecules will be developed by bioengineering methods which may be used for isotope-based diagnosis and treatment of disease.
Collapse
Affiliation(s)
- Uwe Haberkorn
- Department of Nuclear Medicine, University of Heidelberg Clinical Cooperation Unit Nuclear Medicine German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
41
|
Targeting sodium/iodide symporter gene expression for estrogen-regulated imaging and therapy in breast cancer. Cancer Gene Ther 2008; 15:465-73. [PMID: 18421306 DOI: 10.1038/cgt.2008.6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Expression of the sodium iodide symporter (hNIS) has been detected in breast cancer tissue, but frequently, not at the levels necessary to mediate (131)I accumulation. Transducing the hNIS gene into breast cancer cells with adenovirus could be a tractable strategy to render breast cancer susceptible to radioiodide therapy. We constructed the replication-incompetent virus, AdSERE, in which an estrogen-responsive promoter directs the expression of hNIS. In vitro, we demonstrate that AdSERE mediates hNIS expression and iodide uptake in ER+ breast cancer cells. In vivo, we show that AdSERE-infected ER+ tumors can be imaged due to tracer accumulation; in addition, AdSERE in combination with therapeutic doses of (131)I suppresses tumor growth.
Collapse
|
42
|
Spitzweg C, Baker CH, Bergert ER, O'Connor MK, Morris JC. Image-guided radioiodide therapy of medullary thyroid cancer after carcinoembryonic antigen promoter-targeted sodium iodide symporter gene expression. Hum Gene Ther 2007; 18:916-24. [PMID: 17931047 DOI: 10.1089/hum.2007.081] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In contrast to follicular cell-derived thyroid cancer, medullary thyroid cancer (MTC) remains difficult to treat because of its unresponsiveness to radioiodine therapy, or to conventional chemo- and radiotherapy. We therefore examined the feasibility of radioiodine therapy of MTC after human sodium iodide symporter (hNIS) gene transfer, using the tumor-specific carcinoembryonic antigen (CEA) promoter for transcriptional targeting. NIS gene transfer was performed in vivo in human MTC cell (TT) xenografts, using adenoviral vectors carrying the NIS gene linked to the cytomegalovirus promoter (Ad5-CMV-NIS) or a CEA promoter fragment (Ad5-CEA-NIS). Functional NIS expression was confirmed by immunostaining as well as in vivo (123)I gamma-camera imaging followed by application of a therapeutic (131)I dose. TT cell xenografts in nude mice injected intratumorally with Ad5-CEA-NIS accumulated 7.5 +/- 1.2% ID/g (percentage injected dose per gram tumor tissue; 5 x 10(8) PFU) and 12 +/- 2.95% ID/g (1 x 10(9) PFU) with an average biological half-life of 6.1 +/- 0.8 and 23.6 +/- 3.7 hr, respectively, as compared with accumulation of 8.4 +/- 0.9% ID/g with a biological half-life of 12 +/- 8 hr after application of Ad5-CMV-NIS (5 x 10(8) PFU). After Ad5-CEA-NIS-mediated NIS gene transfer in TT cell xenografts administration of a therapeutic dose of 111 MBq (3 mCi) of (131)I resulted in a significant reduction of tumor growth associated with significantly lower calcitonin serum levels in treated mice as well as improved survival. We conclude that a therapeutic effect of (131)I was demonstrated in vivo in MTC cell xenografts after adenovirus-mediated induction of tumor-specific iodide accumulation by CEA promoter-directed hNIS expression.
Collapse
Affiliation(s)
- Christine Spitzweg
- Department of Internal Medicine II, Klinikum Grosshadern, Ludwig-Maximilians-University, 81377 Munich, Germany
| | | | | | | | | |
Collapse
|
43
|
Willhauck MJ, Sharif Samani BR, Gildehaus FJ, Wolf I, Senekowitsch-Schmidtke R, Stark HJ, Göke B, Morris JC, Spitzweg C. Application of 188rhenium as an alternative radionuclide for treatment of prostate cancer after tumor-specific sodium iodide symporter gene expression. J Clin Endocrinol Metab 2007; 92:4451-8. [PMID: 17698909 DOI: 10.1210/jc.2007-0402] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT We reported recently the induction of iodide accumulation in prostate cancer cells (LNCaP) by prostate-specific antigen promoter-directed sodium iodide symporter (NIS) expression that allowed a significant therapeutic effect of (131)iodine ((131)I). These data demonstrated the potential of the NIS gene as a novel therapeutic gene, although in some extrathyroidal tumors, therapeutic efficacy may be limited by rapid iodide efflux due to a lack of iodide organification. OBJECTIVE In the current study, we therefore studied the potential of (188)rhenium ((188)Re), as an alternative radionuclide, also transported by NIS, with a shorter half-life and higher energy beta-particles than (131)I. RESULTS NIS-transfected LNCaP cells (NP-1) concentrated 8% of the total applied activity of (188)Re as compared with 16% of (125)I, which was sufficient for a therapeutic effect in an in vitro clonogenic assay. gamma-Camera imaging of NP-1 cell xenografts in nude mice revealed accumulation of 8-16% injected dose (ID)/g (188)Re (biological half-life 12.9 h), which resulted in a 4.7-fold increased tumor absorbed dose (450 mGy/MBq) for (188)Re as compared with (131)I. After application of 55.5 MBq (131)I or (188)Re, smaller tumors showed a similar average volume reduction of 86%, whereas in larger tumors volume reduction was significantly increased from 73% after (131)I treatment to 85% after application of (188)Re. CONCLUSION Although in smaller prostate cancer xenografts both radionuclides seemed to be equally effective after prostate-specific antigen promoter-mediated NIS gene delivery, a superior therapeutic effect has been demonstrated for (188)Re in larger tumors.
Collapse
Affiliation(s)
- Michael J Willhauck
- Department of Internal Medicine II, Ludwig-Maximilians-University, 81377 Munich, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kanai Y, Hasegawa S, Kimura Y, Oku N, Ito H, Fukuda H, Hatazawa J. N-isopropyl-4-[123I]iodoamphetamine (123I-IMP) products: a difference in radiochemical purity, unmetabolized fraction, and octanol extraction fraction in arterial blood and regional brain uptake in rats. Ann Nucl Med 2007; 21:387-91. [PMID: 17876551 DOI: 10.1007/s12149-007-0038-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 04/13/2007] [Indexed: 11/24/2022]
Abstract
N-isopropyl-4-[123I]iodoamphetamine (123I-IMP) is a lipophilic compound utilized for cerebral blood flow (CBF) measurement with single photon emission computed tomography (SPECT). Two different 123I-IMP products (IMP(A) and IMP(B)) are commercially available. We examined the radiochemical purity, unmetabolized fraction, and octanol extraction fraction in arterial blood, and the regional brain uptake of IMP(A) and IMP(B) in a rat model. IMP(B) (96.4% +/- 0.08%, P < 0.05) showed significantly higher radiochemical purity than IMP(A) (95.5% +/- 0.20%). The mean unmetabolized fraction in arterial blood taken at 10 min after intravenous administration of IMP(B) (69.5% +/- 4.4%, P < 0.01) was significantly higher than that of IMP(A) (59.6% +/- 2.6%). The mean octanol extraction fraction of IMP(B) (75.0% +/- 1.3%, P < 0.01) was also significantly higher than that of IMP(A) (67.2% +/- 0.8%). The mean levels of radioactivity in arterial blood sampled at 10 min after injection and mean regional brain radioactivity (cerebral cortices, basal ganglia, brain stem, and cerebellum) at 10-12 min after injection were not significantly different between IMP(A) and IMP(B). The present study indicates differences in the radiochemical purity and the unmetabolized and octanol extraction fraction in arterial blood between the two commercially available 123I-IMP products. The appropriate octanol extraction fractions for IMP(A) and IMP(B) should be determined in humans and employed for quantitative CBF measurement in clinical SPECT.
Collapse
Affiliation(s)
- Yasukazu Kanai
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Willhauck MJ, Sharif-Samani B, Senekowitsch-Schmidtke R, Wunderlich N, Göke B, Morris JC, Spitzweg C. Functional sodium iodide symporter expression in breast cancer xenografts in vivo after systemic treatment with retinoic acid and dexamethasone. Breast Cancer Res Treat 2007; 109:263-72. [PMID: 17636401 DOI: 10.1007/s10549-007-9646-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Accepted: 06/07/2007] [Indexed: 10/23/2022]
Abstract
CONTEXT The sodium iodide symporter (NIS) mediates iodide uptake in the thyroid gland as well as in lactating breast, and is also expressed in the majority of breast cancers. Recently, we have reported stimulation of all-trans retinoic acid (atRA)-induced NIS expression in the human breast cancer cell line MCF-7 by dexamethasone (Dex), resulting in an enhanced therapeutic effect of (131)I in vitro. OBJECTIVE In the current study we examined the efficacy of Dex stimulation of atRA-induced NIS expression in vivo in MCF-7 xenotransplants in nude mice. DESIGN After systemic treatment with atRA alone or in combination with Dex, iodide accumulation in the tumors was assessed by gamma camera imaging and gamma counter analysis. In addition, NIS expression was examined on RNA and protein level by RT-PCR and immunohistochemistry, respectively. RESULTS Using gamma camera imaging after intraperitoneal injection of 18.5 MBq (123)I, no iodide accumulation was detected in tumors of untreated mice or mice treated with atRA only. After combined treatment with atRA/Dex significant (123)I accumulation was detected in MCF-7 xenografts, which, by ex vivo gamma counting revealed a 3.3-fold increase in iodide accumulation as compared to control tumors. Surprisingly, in a subset of mice treated with atRA or atRA/Dex iodide accumulation was also detected in the normal mammary glands. In a normal human mammary epithelial cell line HB-2, however, no functional NIS expression was induced after treatment with atRA and/or Dex in vitro. Further, NIS mRNA and protein expression was detected in atRA/Dex treated MCF-7 tumors by RT-PCR and immunohistochemistry, respectively. CONCLUSION Treatment with Dex in the presence of atRA is able to induce significant amounts of iodide accumulation in breast cancer xenotransplants in vivo due to stimulation of functional NIS protein expression, which opens exciting perspectives for a possible diagnostic and therapeutic role of radioiodine in the treatment of breast cancer.
Collapse
Affiliation(s)
- Michael J Willhauck
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Schipper ML, Riese CGU, Seitz S, Weber A, Béhé M, Schurrat T, Schramm N, Keil B, Alfke H, Behr TM. Efficacy of 99mTc pertechnetate and 131I radioisotope therapy in sodium/iodide symporter (NIS)-expressing neuroendocrine tumors in vivo. Eur J Nucl Med Mol Imaging 2006; 34:638-650. [PMID: 17160413 DOI: 10.1007/s00259-006-0254-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Accepted: 07/21/2006] [Indexed: 10/23/2022]
Abstract
PURPOSE There is growing interest in the human sodium/iodide symporter (NIS) gene both as a molecular imaging reporter gene and as a therapeutic gene. Here, we show the feasibility of radioisotope therapy of neuroendocrine tumors. As a separate application of NIS gene transfer, we image NIS-expressing tumors with pinhole SPECT in living subjects. METHODS Biodistribution studies and in vivo therapy experiments were performed in nude mice carrying stably NIS-expressing neuroendocrine tumor xenografts following i.v. injection of (131)I and (99m)Tc pertechnetate. To show the usefulness of NIS as an imaging reporter gene, (99m)Tc pertechnetate uptake was imaged in vivo using a clinical gamma camera in combination with a custom-made single pinhole collimator, followed by SPECT/small animal MRI data coregistration. RESULTS NIS-expressing neuroendocrine tumors strongly accumulated (131)I and (99m)Tc pertechnetate, as did thyroid, stomach, and salivary gland. The volume of NIS-expressing neuroendocrine tumors decreased significantly after therapeutic administration of (131)I or (99m)Tc pertechnetate, whereas control tumors continued to grow. NIS-mediated uptake of (99m)Tc pertechnetate could be imaged in vivo at high resolution with a clinical gamma camera equipped with a custom-made single pinhole collimator. High-resolution functional and morphologic information could be combined in a single three-dimensional data set by coregistration of SPECT and small animal MRI data. Lastly, we demonstrated a therapeutic effect of (99m)Tc pertechnetate on NIS-expressing neuroendocrine tumors in cell culture and, for the first time, in vivo, thought to be due to emitted Auger and conversion electrons. CONCLUSIONS NIS-expressing neuroendocrine tumors efficiently concentrate radioisotopes, allowing for in vivo high-resolution small animal SPECT imaging as well as rendering possible successful radioisotope therapy of neuroendocrine tumors.
Collapse
Affiliation(s)
- Meike L Schipper
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany.
- Molecular Imaging Program at Stanford (MIPS) and Department of Radiology, Stanford University, E 150 Clark Center, 318 Campus Drive, Stanford, CA, 94305-5427, USA.
| | - Christoph G U Riese
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| | - Stephan Seitz
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| | - Alexander Weber
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| | - Martin Béhé
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| | - Tino Schurrat
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| | - Nils Schramm
- Department of Electronics, Forschungszentrum Jülich, Jülich, Germany
| | - Boris Keil
- Department of Radiology, Philipps University Marburg, Marburg, Germany
| | - Heiko Alfke
- Department of Radiology, Philipps University Marburg, Marburg, Germany
| | - Thomas M Behr
- Department of Nuclear Medicine, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
47
|
Dayem M, Navarro V, Marsault R, Darcourt J, Lindenthal S, Pourcher T. From the molecular characterization of iodide transporters to the prevention of radioactive iodide exposure. Biochimie 2006; 88:1793-806. [PMID: 16905238 DOI: 10.1016/j.biochi.2006.07.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Accepted: 07/19/2006] [Indexed: 12/23/2022]
Abstract
In the event of a nuclear reactor accident, the major public health risk will likely result from the release and dispersion of volatile radio-iodines. Upon body exposure and food ingestion, these radio-iodines are concentrated in the thyroid, resulting in substantial thyroidal irradiation and accordingly causing thyroid cancers. Stable potassium iodide (KI) effectively blocks thyroid iodine uptake and is thus used in iodide prophylaxis for reactor accidents. The efficiency of KI is directly related to the physiological inhibition of the thyroid function in the presence of high plasma iodide concentrations. This regulation is called the Wolff-Chaikoff effect. However, to be fully effective, KI should be administered shortly before or immediately after radioiodine exposure. If KI is provided only several hours after exposure, it will elicit the opposite effect e.g. lead to an increase in the thyroid irradiation dose. To date, clear evaluation of the benefit and the potential toxicity of KI administration remain difficult, and additional data are needed. We outline in this review the molecular characterization of KI-induced regulation of the thyroid function. Significant advances in the knowledge of the iodide transport mechanisms and thyroid physiology have been made. Recently developed molecular tools should help clarify iodide metabolism and the Wolff-Chaikoff effect. The major goals are clarifying the factors which increase thyroid cancer risk after a reactor accident and improving the KI administration protocol. These will ultimately lead to the development of novel strategies to decrease thyroid irradiation after radio-iodine exposure.
Collapse
Affiliation(s)
- M Dayem
- Unité TIRO (Transporter in Imaging and Radiotherapy in Oncology), Commissariat à l'énergie atomique DSV-DIEP-SBTN, School of Medicine, University of Nice Sophia Antipolis, 28, avenue de Valombrose, 06107 Nice cedex, France
| | | | | | | | | | | |
Collapse
|
48
|
Ito H, Sato T, Odagiri H, Inoue K, Shidahara M, Suhara T, Hatazawa J, Fukuda H. Brain and whole body distribution ofN-isopropyl-4-iodoamphetamine (I-123) in humans: Comparison of radiopharmaceuticals marketed by different companies in Japan. Ann Nucl Med 2006; 20:493-8. [PMID: 17037282 DOI: 10.1007/bf02987259] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Iodine-123 (123I)-labeled N-isopropyl-4-iodoamphetamine (IMP) has been used as a cerebral blood flow (CBF) tracer for single-photon emission computed tomography (SPECT). An autoradiographic (ARG) method has been developed for the quantitation of CBF by IMP and SPECT. Two IMPs (IMPA and IMPB) produced by different radiopharmaceutical companies are marketed in Japan. In the present study, whole-body distributions including brain and blood of the two IMPs were compared in the same human subjects. METHODS Two brain SPECT studies using IMPA or IMPB were performed on separate days in six young healthy men. Whole-body scans were also obtained with a large field-of-view single-head gamma camera. One-point arterial blood sampling was performed at 10 min after injection of IMP to measure both the radioactivity concentrations of whole blood and of octanol-extracted components. RESULTS No significant differences between the two tracers were observed in body distribution, tracer kinetics in brain, or regional distribution in brain. However, the octanol extraction fraction in blood was significantly different between the two tracers. Radiochemical purity was slightly but significantly different between the tracers. CONCLUSIONS In the ARG method, arterial input function is determined by calibration of a standard input function with the radioactivity concentration of arterial whole blood. Because the standard input function in the ARG method was obtained using IMPA, the standard input function obtained for IMPB should be used when CBF is calculated by the ARG method with IMPB.
Collapse
Affiliation(s)
- Hiroshi Ito
- Department of Nuclear Medicine and Radiology, Division of Brain Sciences, Institute of Development, Aging and Cancer, Tohoku University.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Dadachova E, Nguyen A, Lin EY, Gnatovskiy L, Lu P, Pollard JW. Treatment with rhenium-188-perrhenate and iodine-131 of NIS-expressing mammary cancer in a mouse model remarkably inhibited tumor growth. Nucl Med Biol 2005; 32:695-700. [PMID: 16243644 DOI: 10.1016/j.nucmedbio.2005.05.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 05/13/2005] [Accepted: 05/15/2005] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Novel therapeutic modalities are needed for breast cancer patients in whom standard treatments are not effective. Mammary gland sodium/iodide symporter has been identified as a molecular target in breast cancers in humans and in some transgenic mouse models. We report the results of a therapy study with (131)I(-) and (188)ReO(4)(-) of breast cancer in polyoma middle T oncoprotein (PyMT) transgenic mice endogenously expressing the Na(+)/I(-) symporter (NIS). METHODS PyMT mice (12-13 weeks old) with one palpable tumor of 0.5-0.8 cm in diameter were used. For the therapy studies, PyMT mice were (1) treated with two intraperitoneal injections of 1.5 mCi of (188)ReO(4)(-) 1 week apart, (2) pretreated for 1 week with 5 microg of triiodothyronine (T3) followed by two intraperitoneal injections of 1.5 mCi of (131)I(-) 1 week apart or (3) left untreated. The tumor and normal organ uptakes were assessed by scintigraphic imaging. The thyroid function of treated and control animals was evaluated at the completion of the study by measuring the T3/thyroxine (T4) ratio in their blood. RESULTS There was significant uptake of (131)I(-) and (188)ReO(4)(-) in the primary palpable tumors as well as in nonpalpable tumors, stomachs and thyroids. The tumor uptake after the second injection was 10 times lower in comparison with the first injection. Tumor growth was significantly inhibited in both the (131)I(-) and (188)ReO(4)(-) groups in comparison with the control group, and tumors in the (188)ReO(4)(-) group increased in size significantly less than in the (131)I(-) group. The T3/T4 ratios were calculated to be 27 and 25 for the control group and the (188)ReO(4)(-) group, respectively; for (131)I(-), both the T3 and T4 levels were below detection limit, demonstrating much less effect on the thyroids of treatment with (188)ReO(4)(-) than with (131)I(-). CONCLUSIONS These results prove that NIS expression in breast tumors in animal models allows specific, efficient and safe treatment with a variety of radionuclides transported by NIS.
Collapse
Affiliation(s)
- Ekaterina Dadachova
- Department of Nuclear Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Oh HJ, Chung JK, Kang JH, Kang WJ, Noh DY, Park IA, Jeong JM, Lee DS, Lee MC. The relationship between expression of the sodium/iodide symporter gene and the status of hormonal receptors in human breast cancer tissue. Cancer Res Treat 2005; 37:247-50. [PMID: 19956522 DOI: 10.4143/crt.2005.37.4.247] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Accepted: 07/02/2005] [Indexed: 11/21/2022] Open
Abstract
PURPOSE It has been reported that the sodium/iodide symporter (NIS) gene is expressed in several breast cancer tissues, suggesting the possibility of radionuclide imaging and therapy. However, the regulatory mechanism of NIS gene expression in breast cancer is not yet understood. To assess the relationship between the hormonal status and the NIS expression in breast cancer tissue, we investigated the NIS expression and correlated it to the expression of the thyrotropin receptor (thyroid stimulating hormone receptor, TSH-R), the estrogen receptor (ER) and the progesterone receptor (PR) in human breast cancer tissues. MATERIALS AND METHODS Breast cancer tissues were obtained from 44 patients. Pathological examination showed 2 cases of Grade I, 17 of Grade II, 22 of Grade III, and 3 of unknown grade. We measured the expression of NIS and TSH-R genes by using RT-PCR and we measured the status of ER and PR by using immunohistochemistry. RESULTS The NIS gene was expressed in 15 (34%) of the 44 breast cancer tissues. The NIS gene was expressed in 32% of the cases with TSH-R gene expression. The NIS gene was expressed in 40% of the breast cancer tissues with a positive PR and in 31% with a negative PR (p>0.05). It was positive for PR in 18% of the cases and negative for PR in 39% of the cases (p>0.05). CONCLUSION The NIS gene is expressed in approximately one-third of the human breast cancer tissues. Its expression was not related to the presence of the TSH-R gene or hormonal receptors, ER and PR.
Collapse
Affiliation(s)
- Hyun Jung Oh
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|