1
|
Shalaby N, Xia Y, Kelly JJ, Sanchez-Pupo R, Martinez F, Fox MS, Thiessen JD, Hicks JW, Scholl TJ, Ronald JA. Imaging CAR-NK cells targeted to HER2 ovarian cancer with human sodium-iodide symporter-based positron emission tomography. Eur J Nucl Med Mol Imaging 2024; 51:3176-3190. [PMID: 38722382 PMCID: PMC11368970 DOI: 10.1007/s00259-024-06722-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/14/2024] [Indexed: 09/03/2024]
Abstract
Chimeric antigen receptor (CAR) cell therapies utilize CARs to redirect immune cells towards cancer cells expressing specific antigens like human epidermal growth factor receptor 2 (HER2). Despite their potential, CAR T cell therapies exhibit variable response rates and adverse effects in some patients. Non-invasive molecular imaging can aid in predicting patient outcomes by tracking infused cells post-administration. CAR-T cells are typically autologous, increasing manufacturing complexity and costs. An alternative approach involves developing CAR natural killer (CAR-NK) cells as an off-the-shelf allogeneic product. In this study, we engineered HER2-targeted CAR-NK cells co-expressing the positron emission tomography (PET) reporter gene human sodium-iodide symporter (NIS) and assessed their therapeutic efficacy and PET imaging capability in a HER2 ovarian cancer mouse model.NK-92 cells were genetically modified to express a HER2-targeted CAR, the bioluminescence imaging reporter Antares, and NIS. HER2-expressing ovarian cancer cells were engineered to express the bioluminescence reporter Firefly luciferase (Fluc). Co-culture experiments demonstrated significantly enhanced cytotoxicity of CAR-NK cells compared to naive NK cells. In vivo studies involving mice with Fluc-expressing tumors revealed that those treated with CAR-NK cells exhibited reduced tumor burden and prolonged survival compared to controls. Longitudinal bioluminescence imaging demonstrated stable signals from CAR-NK cells over time. PET imaging using the NIS-targeted tracer 18F-tetrafluoroborate ([18F]TFB) showed significantly higher PET signals in mice treated with NIS-expressing CAR-NK cells.Overall, our study showcases the therapeutic potential of HER2-targeted CAR-NK cells in an aggressive ovarian cancer model and underscores the feasibility of using human-derived PET reporter gene imaging to monitor these cells non-invasively in patients.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Ying Xia
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - John J Kelly
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Rafael Sanchez-Pupo
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Francisco Martinez
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew S Fox
- Lawson Health Research Institute, London, ON, Canada
- Saint Joseph's Health Care, London, ON, Canada
| | - Jonathan D Thiessen
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, ON, Canada
- Saint Joseph's Health Care, London, ON, Canada
| | - Justin W Hicks
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, ON, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Ontario Institute for Cancer Research, London, ON, Canada
| | - John A Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
2
|
Oh SG, Choi JY, Lee JE, Jeon S, Lee BR, Son KH, Lee SB, An BS, Hwang DY, Kim SJ, Ha KT, Lee J, Jeon YH. Visualizing mast cell migration to tumor sites using sodium iodide symporter of nuclear medicine reporter gene. Neoplasia 2023; 43:100925. [PMID: 37562258 PMCID: PMC10423699 DOI: 10.1016/j.neo.2023.100925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
PURPOSE Owing to the close relationship between mast cells and cancer progression, an imaging technique that can be applied in a clinical setting to explore the biological behavior of mast cells in the tumor microenvironment is needed. In this study, we visualized mast cell migration to lung tumor lesions in live mice using sodium iodide symporter (NIS) as a nuclear medicine reporter gene. EXPERIMENTAL DESIGN The murine mast cell line MC-9 was infected with retrovirus including NIS, luciferase (as a surrogate marker for NIS), and Thy1.1 to generate MC-9/NFT cells. Radioiodine uptake was measured in MC-9/NFT cells, and an inhibition assay of radioiodine uptake using KCLO4 was also performed. Cell proliferation and FcεRI expression was examined in MC-9 and MC-9/NFT cells. The effect of mast cell-conditioned media (CM) on the proliferation of Lewis lung cancer (LLC) cells was examined. The migration level of MC-9/NFT cells was confirmed in the presence of serum-free media (SFM) and CM of cancer cells. After intravenous injection of MC-9/NFT cells into mice with an LLC tumor, I-124 PET/CT and biodistribution analysis was performed. RESULTS MC-9/NFT cells exhibited higher radioiodine avidity compared to parental MC-9 cells; this increased radioiodine avidity in MC-9/NFT cells was reduced to basal level by KCLO4. Levels of FcεRI expression and cell proliferation were not different in parental MC-9 cell and MC-9/ NFT cells. The CM of MC-9/NFT cells increased cancer cell proliferation relative to that of the SFM. The migration level of MC-9/NFT cells was higher in the CM than the SFM of LLC cells. PET/CT imaging with I-124 clearly showed infiltration of reporter mast cells in lung tumor at 24 h after transfer, which was consistent with the findings of the biodistribution examination. CONCLUSION These findings suggest that the sodium iodide symporter can serve as a reliable nuclear medicine reporter gene for non-invasively imaging the biological activity of mast cells in mice with lung tumors. Visualizing mast cells in the tumor microenvironment via a nuclear medicine reporter gene would provide valuable insights into their biological functions.
Collapse
Affiliation(s)
- Seul-Gi Oh
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea; Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun Young Choi
- Preclincial Research Center (PRC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea; Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Jae-Eon Lee
- Preclincial Research Center (PRC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea; Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - SoYeon Jeon
- Preclincial Research Center (PRC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea
| | - Bo-Ra Lee
- Preclincial Research Center (PRC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea
| | - Kwang Hee Son
- Preclincial Research Center (PRC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea
| | - Sang Bong Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Seong-Jang Kim
- Pusan National University College of Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Ki-Tae Ha
- Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yong Hyun Jeon
- Preclincial Research Center (PRC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Republic of Korea.
| |
Collapse
|
3
|
Shao Y, Wang Y, Su R, Pu W, Chen S, Fu L, Yu H, Qiu Y. Dual identity of tumor-associated macrophage in regulated cell death and oncotherapy. Heliyon 2023; 9:e17582. [PMID: 37449180 PMCID: PMC10336529 DOI: 10.1016/j.heliyon.2023.e17582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/25/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023] Open
Abstract
Tumor-associated macrophage (TAM) affects the intrinsic properties of tumor cells and the tumor microenvironment (TME), which can stimulate tumor cell proliferation, migration, and genetic instability, and macrophage diversity includes the diversity of tumors with different functional characteristics. Macrophages are now a central drug target in various diseases, especially in the TME, which, as "tumor promoters" and "immunosuppressors", have different responsibilities during tumor development and accompany by significant dynamic alterations in various subpopulations. Remodelling immunosuppression of TME and promotion of pre-existing antitumor immune responses is critical by altering TAM polarization, which is relevant to the efficacy of immunotherapy, and uncovering the exact mechanism of action of TAMs and identifying their specific targets is vital to optimizing current immunotherapies. Hence, this review aims to reveal the triadic interactions of macrophages with programmed death and oncotherapy, and to integrate certain relationships in cancer treatment.
Collapse
Affiliation(s)
- Yingying Shao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yu Wang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Ranran Su
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Weiling Pu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen, China
- Department of Applied Biology and Chemical Technology, Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, China
| | - Leilei Fu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Haiyang Yu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Reporter Genes for Brain Imaging Using MRI, SPECT and PET. Int J Mol Sci 2022; 23:ijms23158443. [PMID: 35955578 PMCID: PMC9368793 DOI: 10.3390/ijms23158443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023] Open
Abstract
The use of molecular imaging technologies for brain imaging can not only play an important supporting role in disease diagnosis and treatment but can also be used to deeply study brain functions. Recently, with the support of reporter gene technology, optical imaging has achieved a breakthrough in brain function studies at the molecular level. Reporter gene technology based on traditional clinical imaging modalities is also expanding. By benefiting from the deeper imaging depths and wider imaging ranges now possible, these methods have led to breakthroughs in preclinical and clinical research. This article focuses on the applications of magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), and positron emission tomography (PET) reporter gene technologies for use in brain imaging. The tracking of cell therapies and gene therapies is the most successful and widely used application of these techniques. Meanwhile, breakthroughs have been achieved in the research and development of reporter genes and their imaging probe pairs with respect to brain function research. This paper introduces the imaging principles and classifications of the reporter gene technologies of these imaging modalities, lists the relevant brain imaging applications, reviews their characteristics, and discusses the opportunities and challenges faced by clinical imaging modalities based on reporter gene technology. The conclusion is provided in the last section.
Collapse
|
5
|
Shao F, Long Y, Ji H, Jiang D, Lei P, Lan X. Radionuclide-based molecular imaging allows CAR-T cellular visualization and therapeutic monitoring. Am J Cancer Res 2021; 11:6800-6817. [PMID: 34093854 PMCID: PMC8171102 DOI: 10.7150/thno.56989] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is a new and effective form of adoptive cell therapy that is rapidly entering the mainstream for the treatment of CD19-positive hematological cancers because of its impressive effect and durable responses. Huge challenges remain in achieving similar success in patients with solid tumors. The current methods of monitoring CAR-T, including morphological imaging (CT and MRI), blood tests, and biopsy, have limitations to assess whether CAR-T cells are homing to tumor sites and infiltrating into tumor bed, or to assess the survival, proliferation, and persistence of CAR-T cells in solid tumors associated with an immunosuppressive microenvironment. Radionuclide-based molecular imaging affords improved CAR-T cellular visualization and therapeutic monitoring through either a direct cellular radiolabeling approach or a reporter gene imaging strategy, and endogenous cell imaging is beneficial to reflect functional information and immune status of T cells. Focusing on the dynamic monitoring and precise assessment of CAR-T therapy, this review summarizes the current applications of radionuclide-based noninvasive imaging in CAR-T cells visualization and monitoring and presents current challenges and strategic choices.
Collapse
|
6
|
Miederer M, Pektor S, Miederer I, Bausbacher N, Keil IS, Hefesha H, Haas H, Sahin U, Diken M. Iodine-124 PET quantification of organ-specific delivery and expression of NIS-encoding RNA. EJNMMI Res 2021; 11:14. [PMID: 33569663 PMCID: PMC7876195 DOI: 10.1186/s13550-021-00753-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/01/2021] [Indexed: 11/19/2022] Open
Abstract
Background RNA-based vaccination strategies tailoring immune response to specific reactions have become an important pillar for a broad range of applications. Recently, the use of lipid-based nanoparticles opened the possibility to deliver RNA to specific sites within the body, overcoming the limitation of rapid degradation in the bloodstream. Here, we have investigated whether small animal PET/MRI can be employed to image the biodistribution of RNA-encoded protein.
For this purpose, a reporter RNA coding for the sodium-iodide-symporter (NIS) was in vitro transcribed in cell lines and evaluated for expression. RNA-lipoplex nanoparticles were then assembled by complexing RNA with liposomes at different charge ratios, and functional NIS protein translation was imaged and quantified in vivo and ex vivo by Iodine-124 PET upon intravenous administration in mice. Results NIS expression was detected on the membrane of two cell lines as early as 6 h after transfection and gradually decreased over 48 h. In vivo and ex vivo PET/MRI of anionic spleen-targeting or cationic lung-targeting NIS-RNA lipoplexes revealed a visually detectable rapid increase of Iodine-124 uptake in the spleen or lung compared to control-RNA-lipoplexes, respectively, with minimal background in other organs except from thyroid, stomach and salivary gland. Conclusions The strong organ selectivity and high target-to-background acquisition of NIS-RNA lipoplexes indicate the feasibility of small animal PET/MRI to quantify organ-specific delivery of RNA. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00753-2.
Collapse
Affiliation(s)
- Matthias Miederer
- Department of Nuclear Medicine, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Stefanie Pektor
- Department of Nuclear Medicine, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Isabelle Miederer
- Department of Nuclear Medicine, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Nicole Bausbacher
- Department of Nuclear Medicine, University Medical Center of Johannes Gutenberg University, Mainz, Germany
| | - Isabell Sofia Keil
- TRON - Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Hossam Hefesha
- Biopharmaceutical New Technologies (BioNTech) SE, Mainz, Germany
| | - Heinrich Haas
- Biopharmaceutical New Technologies (BioNTech) SE, Mainz, Germany
| | - Ugur Sahin
- TRON - Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz gGmbH, Mainz, Germany.,Biopharmaceutical New Technologies (BioNTech) SE, Mainz, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz gGmbH, Mainz, Germany. .,Biopharmaceutical New Technologies (BioNTech) SE, Mainz, Germany.
| |
Collapse
|
7
|
Jacobs AH, Schelhaas S, Viel T, Waerzeggers Y, Winkeler A, Zinnhardt B, Gelovani J. Imaging of Gene and Cell-Based Therapies: Basis and Clinical Trials. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
8
|
In vivo cell tracking with viral vector mediated genetic labeling. J Neurosci Methods 2020; 350:109021. [PMID: 33316318 DOI: 10.1016/j.jneumeth.2020.109021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Cell tracking is a useful technique to monitor specific cell populations for their morphology, development, proliferation, migration, interaction, function, and other properties, both in vitro and in vivo. Using different materials and methodologies to label the target cells directly or indirectly, the dynamic biological processes in living organisms can be visualized with appropriate detection techniques. Viruses, with the unique ability to deliver exogenous genes into host cells, have been used as vectors to mediate gene transfer. Genetic labeling of target cells by viral vectors endows the cells to express reporter genes with high efficiency and specificity. In conjunction with corresponding imaging techniques, cells labeled with different genetic reporters mediated by different viral vectors can be monitored across spatial and temporal scales to fulfill various purposes and address different questions. In the present review, we introduce the basic principle of viral vectors in cell tracking and highlight the examples of cell tracking in various research areas.
Collapse
|
9
|
Bu L, Sun Y, Han G, Tu N, Xiao J, Wang Q. Outcome Prediction and Evaluation by Imaging the Key Elements of Therapeutic Responses to Cancer Immunotherapies Using PET. Curr Pharm Des 2020; 26:675-687. [PMID: 31465273 DOI: 10.2174/1381612825666190829150302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/21/2019] [Indexed: 12/23/2022]
Abstract
Cancer immunotherapy (also known as immuno-oncology), a promising anti-cancer strategy by harnessing the body's own immune system against cancer, has emerged as the "fifth therapeutic pilla" in the field of cancer treatment since surgery, chemotherapy, radiation and targeted therapy. Clinical efficacy of several immunotherapies has been demonstrated in clinical settings, however, only a small subset of patients exhibit dramatic or durable responses, with the highest reported frequency about 10-40% from single-agent PD-L1/PD-1 inhibitors, suggesting the urgent need of consistent objective response biomarkers for monitoring therapeutic response accurately, predicting therapeutic efficacy and selecting responders. Key elements of therapeutic responses to cancer immunotherapies contain the cancer cell response and the alternation of inherent immunological characteristics. Here, we document the literature regarding imaging the key elements of therapeutic responses to cancer immunotherapies using PET. We discussed PET imaging approaches according to different response mechanisms underlying diverse immune-therapeutic categories, and also highlight the ongoing efforts to identify novel immunotherapeutic PET imaging biomarkers. In this article, we show that PET imaging of the key elements of therapeutic responses to cancer immunotherapies using PET can allow for more precise prediction, earlier therapy response monitoring, and improved management. However, all of these strategies need more preclinical study and clinical validation before further development as imaging indicators of the immune response.
Collapse
Affiliation(s)
- Lihong Bu
- PET-CT/MRI Center, Faculty of Radiology and Nuclear Medicine, Wuhan University Renmin Hospital, Wuhan, Hubei, China
| | - Yanqiu Sun
- Department of Radiology, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Guang Han
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ning Tu
- PET-CT/MRI Center, Faculty of Radiology and Nuclear Medicine, Wuhan University Renmin Hospital, Wuhan, Hubei, China
| | - Jiachao Xiao
- PET-CT/MRI Center, Faculty of Radiology and Nuclear Medicine, Wuhan University Renmin Hospital, Wuhan, Hubei, China
| | - Qi Wang
- The 1st Department of Gastrointestinal Surgery, Wuhan University Renmin Hospital, Wuhan, Hubei, China
| |
Collapse
|
10
|
Concilio SC, Suksanpaisan L, Pham L, Peng KW, Russell SJ. Improved Noninvasive In Vivo Tracking of AAV-9 Gene Therapy Using the Perchlorate-Resistant Sodium Iodide Symporter from Minke Whale. Mol Ther 2020; 29:236-243. [PMID: 33038323 PMCID: PMC7791078 DOI: 10.1016/j.ymthe.2020.09.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 01/12/2023] Open
Abstract
The sodium iodide symporter (NIS) is widely used as a reporter gene to noninvasively monitor the biodistribution and durability of vector-mediated gene expression via gamma scintigraphy, single-photon emission computed tomography (SPECT), and positron-emission tomography (PET). However, the approach is limited by background signal due to radiotracer uptake by endogenous NIS-expressing tissues. In this study, using the SPECT tracer pertechnetate (99mTcO4) and the PET tracer tetrafluoroborate (B18F4), in combination with the NIS inhibitor perchlorate, we compared the transport properties of human NIS and minke whale (Balaenoptera acutorostrata scammoni) NIS in vitro and in vivo. Based on its relative resistance to perchlorate, the NIS protein from minke whale appeared to be the superior candidate reporter gene. SPECT and PET imaging studies in nude mice challenged with NIS-encoding adeno-associated virus (AAV)-9 vectors confirmed that minke whale NIS, in contrast to human and endogenous mouse NIS, continues to function as a reliable reporter even when background radiotracer uptake by endogenous NIS is blocked by perchlorate.
Collapse
Affiliation(s)
- Susanna C Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Linh Pham
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA.
| |
Collapse
|
11
|
Dual-engineered, “Trojanized” macrophages bio-modally eradicate tumors through biologically and photothermally deconstructing cancer cells in an on-demand, NIR-commanded, self-explosive manner. Biomaterials 2020; 250:120021. [DOI: 10.1016/j.biomaterials.2020.120021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 12/20/2022]
|
12
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
13
|
Reporter gene imaging and its role in imaging-based drug development. Drug Discov Today 2020; 25:582-592. [DOI: 10.1016/j.drudis.2019.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/20/2019] [Accepted: 12/31/2019] [Indexed: 01/21/2023]
|
14
|
Enhanced noninvasive imaging of oncology models using the NIS reporter gene and bioluminescence imaging. Cancer Gene Ther 2019; 27:179-188. [PMID: 30674994 PMCID: PMC7170803 DOI: 10.1038/s41417-019-0081-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/11/2018] [Accepted: 12/28/2018] [Indexed: 02/08/2023]
Abstract
Noninvasive bioluminescence imaging (BLI) of luciferase-expressing tumor cells has advanced pre-clinical evaluation of cancer therapies. Yet despite its successes, BLI is limited by poor spatial resolution and signal penetration, making it unusable for deep tissue or large animal imaging and preventing precise anatomical localization or signal quantification. To refine pre-clinical BLI methods and circumvent these limitations, we compared and ultimately combined BLI with tomographic, quantitative imaging of the sodium iodide symporter (NIS). To this end, we generated tumor cell lines expressing luciferase, NIS, or both reporters, and established tumor models in mice. BLI provided sensitive early detection of tumors and relatively easy monitoring of disease progression. However, spatial resolution was poor, and as the tumors grew, deep thoracic tumor signals were massked by overwhelming surface signals from superficial tumors. In contrast, NIS-expressing tumors were readily distinguished and precisely localized at all tissue depths by positron emission tomography (PET) or single photon emission computed tomography (SPECT) imaging. Furthermore, radiotracer uptake for each tumor could be quantitated noninvasively. Ultimately, combining BLI and NIS imaging represented a significant enhancement over traditional BLI, providing more information about tumor size and location. This combined imaging approach should facilitate comprehensive evaluation of tumor responses to given therapies.
Collapse
|
15
|
Abe T, Sumi K, Kunimatsu R, Oki N, Tsuka Y, Nakajima K, Tanimoto K. Dynamic imaging of the effect of mesenchymal stem cells on osteoclast precursor cell chemotaxis for bone defects in the mouse skull. J Dent Sci 2018; 13:354-359. [PMID: 30895145 PMCID: PMC6388849 DOI: 10.1016/j.jds.2018.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/20/2018] [Indexed: 01/08/2023] Open
Abstract
Background/purpose Mesenchymal stem cells (MSCs) transplantation has previously been used in the field of regenerative medicine. Although bone regeneration is known to occur through the interaction between osteoblasts and osteoclasts, the effect of MSCs on osteoclasts is unknown. Therefore, the purpose of this study was to investigate the effect of MSCs on the chemotaxis of osteoclast precursor cells (RAW264 macrophage cells). Materials and methods Bone defects were created in mice skulls, and MSCs and a scaffold of carbonated hydroxyapatite were transplanted into the bone defects. RAW264 cells were then transplanted into the mouse tail vein, and their dynamics were observed by an in vivo imaging system. Results The fluorescent intensity of the MSCs transplant group at the bone defect region was significantly higher on days 3, 5, and 7 compared with the MSCs non-transplant group. Conclusion Increased RAW264 chemotaxis to the bone defect region occurred following the simultaneous implantation of MSCs in the skull defect.
Collapse
Affiliation(s)
- Takaharu Abe
- Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, Japan
| | - Keisuke Sumi
- Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, Japan
| | - Ryo Kunimatsu
- Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, Japan
| | - Nanae Oki
- Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, Japan
| | - Yuji Tsuka
- Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, Japan
| | - Kengo Nakajima
- Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical & Health Sciences, Japan
| |
Collapse
|
16
|
Abstract
The recent clinical success of cancer immunotherapy has renewed interest in the development of tools to image the immune system. In general, immunotherapies attempt to enable the body's own immune cells to seek out and destroy malignant disease. Molecular imaging of the cells and molecules that regulate immunity could provide unique insight into the mechanisms of action, and failure, of immunotherapies. In this article, we will provide a comprehensive overview of the current state-of-the-art immunoimaging toolbox with a focus on imaging strategies and their applications toward immunotherapy.
Collapse
Affiliation(s)
- Aaron T Mayer
- Department of Bioengineering, Stanford University, Stanford, California; and
| | - Sanjiv S Gambhir
- Department of Bioengineering, Stanford University, Stanford, California; and
- Department of Radiology, Department of Materials Science and Engineering, Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| |
Collapse
|
17
|
Emami-Shahri N, Foster J, Kashani R, Gazinska P, Cook C, Sosabowski J, Maher J, Papa S. Clinically compliant spatial and temporal imaging of chimeric antigen receptor T-cells. Nat Commun 2018. [PMID: 29540684 PMCID: PMC5852048 DOI: 10.1038/s41467-018-03524-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The unprecedented efficacy of chimeric antigen receptor (CAR) T-cell immunotherapy of CD19+ B-cell malignancy has established a new therapeutic pillar of hematology–oncology. Nonetheless, formidable challenges remain for the attainment of comparable success in patients with solid tumors. To accelerate progress and rapidly characterize emerging toxicities, systems that permit the repeated and non-invasive assessment of CAR T-cell bio-distribution would be invaluable. An ideal solution would entail the use of a non-immunogenic reporter that mediates specific uptake of an inexpensive, non-toxic and clinically established imaging tracer by CAR T cells. Here we show the utility of the human sodium iodide symporter (hNIS) for the temporal and spatial monitoring of CAR T-cell behavior in a cancer-bearing host. This system provides a clinically compliant toolkit for high-resolution serial imaging of CAR T cells in vivo, addressing a fundamental unmet need for future clinical development in the field. Adoptive transfer of chimeric antigen receptor (CAR) T cells has shown promising anticancer results in clinical trials. Here the authors use the human sodium iodide symporter (hNIS) as a reporter gene to image human CAR T cells in cancer-bearing mice using broadly available tracers and imaging platforms.
Collapse
Affiliation(s)
- Nia Emami-Shahri
- ImmunoEngineering Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK
| | - Julie Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Roxana Kashani
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Patrycja Gazinska
- Breast Cancer Now, Division of Cancer Studies, Guy's Cancer Centre, Great Maze Pond, London, SE1 9RT, UK
| | - Celia Cook
- ImmunoEngineering Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - John Maher
- CAR Mechanics Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK.,Department of Immunology, Eastbourne Hospital, King's Drive, Eastbourne, BN21 2UD, UK
| | - Sophie Papa
- ImmunoEngineering Group, King's College London, Division of Cancer Studies, 3rd Floor Bermondsey Wing, King's Health Partners Integrated Cancer Centre, Great Maze Pond, Guy's Hospital, London, SE1 9RT, UK. .,Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
18
|
Volpe A, Man F, Lim L, Khoshnevisan A, Blower J, Blower PJ, Fruhwirth GO. Radionuclide-fluorescence Reporter Gene Imaging to Track Tumor Progression in Rodent Tumor Models. J Vis Exp 2018:57088. [PMID: 29608157 PMCID: PMC5931757 DOI: 10.3791/57088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Metastasis is responsible for most cancer deaths. Despite extensive research, the mechanistic understanding of the complex processes governing metastasis remains incomplete. In vivo models are paramount for metastasis research, but require refinement. Tracking spontaneous metastasis by non-invasive in vivo imaging is now possible, but remains challenging as it requires long-time observation and high sensitivity. We describe a longitudinal combined radionuclide and fluorescence whole-body in vivo imaging approach for tracking tumor progression and spontaneous metastasis. This reporter gene methodology employs the sodium iodide symporter (NIS) fused to a fluorescent protein (FP). Cancer cells are engineered to stably express NIS-FP followed by selection based on fluorescence-activated cell sorting. Corresponding tumor models are established in mice. NIS-FP expressing cancer cells are tracked non-invasively in vivo at the whole-body level by positron emission tomography (PET) using the NIS radiotracer [18F]BF4-. PET is currently the most sensitive in vivo imaging technology available at this scale and enables reliable and absolute quantification. Current methods either rely on large cohorts of animals that are euthanized for metastasis assessment at varying time points, or rely on barely quantifiable 2D imaging. The advantages of the described method are: (i) highly sensitive non-invasive in vivo 3D PET imaging and quantification, (ii) automated PET tracer production, (iii) a significant reduction in required animal numbers due to repeat imaging options, (iv) the acquisition of paired data from subsequent imaging sessions providing better statistical data, and (v) the intrinsic option for ex vivo confirmation of cancer cells in tissues by fluorescence microscopy or cytometry. In this protocol, we describe all steps required for routine NIS-FP-afforded non-invasive in vivo cancer cell tracking using PET/CT and ex vivo confirmation of in vivo results. This protocol has applications beyond cancer research whenever in vivo localization, expansion and long-time monitoring of a cell population is of interest.
Collapse
Affiliation(s)
- Alessia Volpe
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Francis Man
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Lindsay Lim
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Alex Khoshnevisan
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Julia Blower
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Philip J Blower
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London
| | - Gilbert O Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London;
| |
Collapse
|
19
|
Drug Discovery by Molecular Imaging and Monitoring Therapy Response in Lymphoma. Int J Mol Sci 2017; 18:ijms18081639. [PMID: 28749424 PMCID: PMC5578029 DOI: 10.3390/ijms18081639] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/23/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022] Open
Abstract
Molecular imaging allows a noninvasive assessment of biochemical and biological processes in living subjects. Treatment strategies for malignant lymphoma depend on histology and tumor stage. For the last two decades, molecular imaging has been the mainstay diagnostic test for the staging of malignant lymphoma and the assessment of response to treatment. This technology enhances our understanding of disease and drug activity during preclinical and clinical drug development. Here, we review molecular imaging applications in drug development, with an emphasis on oncology. Monitoring and assessing the efficacy of anti-cancer therapies in preclinical or clinical models are essential and the multimodal molecular imaging approach may represent a new stage for pharmacologic development in cancer. Monitoring the progress of lymphoma therapy with imaging modalities will help patients. Identifying and addressing key challenges is essential for successful integration of molecular imaging into the drug development process. In this review, we highlight the general usefulness of molecular imaging in drug development and radionuclide-based reporter genes. Further, we discuss the different molecular imaging modalities for lymphoma therapy and their preclinical and clinical applications.
Collapse
|
20
|
Ahn SB, Lee SB, Singh TD, Cho SJ, Kim SK, Lee IK, Jeong SY, Ahn BC, Lee J, Lee SW, Jeon YH. Multimodality Imaging of Bone Marrow-Derived Dendritic Cell Migration and Antitumor Immunity. Transl Oncol 2017; 10:262-270. [PMID: 28214774 PMCID: PMC5314440 DOI: 10.1016/j.tranon.2017.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 11/17/2022] Open
Abstract
Here, we sought to monitor bone marrow-derived dendritic cell (BMDC) migration and antitumor effects using a multimodal reporter imaging strategy in living mice. BMDCs were transduced with retroviral vector harboring human sodium iodide symporter (hNIS, nuclear imaging reporter), firefly luc2 (optical imaging reporter), and thy1.1 (surrogate marker of NIS and luc2) genes (BMDC/NF cells). No significant differences in biological functions, including cell proliferation, antigen uptake, phenotype expression, and migration ability, were observed between BMDC and BMDC/NF cells. Combined bioluminescence imaging and I-124 positron emission tomography/computed tomography clearly revealed the migration of BMDC/NF cells to draining popliteal lymph nodes at day 7 postinjection. Interestingly, marked tumor protection was observed in mice immunized with TC-1 lysate-pulsed BMDC/NF cells. Our findings suggested that multimodal reporter gene imaging of NIS and luciferase could provide insights into the biological behaviors of dendritic cells in living organisms and could be a useful tool for the optimization of DC-based immunotherapy protocols.
Collapse
Affiliation(s)
- Su-Bi Ahn
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Sang Bong Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Thoudam Debraj Singh
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Sung Jin Cho
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, South Korea
| | - Sang Kyoon Kim
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea; Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
| | - Yong Hyun Jeon
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea; Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea.
| |
Collapse
|
21
|
A Novel Ideal Radionuclide Imaging System for Non-invasively Cell Monitoring built on Baculovirus Backbone by Introducing Sleeping Beauty Transposon. Sci Rep 2017; 7:43879. [PMID: 28262785 PMCID: PMC5338331 DOI: 10.1038/srep43879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/30/2017] [Indexed: 12/17/2022] Open
Abstract
Sleeping Beauty (SB) transposon is an attractive tool in stable transgene integration both in vitro and in vivo; and we introduced SB transposon into recombinant sodium-iodide symporter baculovirus system (Bac-NIS system) to facilitate long-term expression of recombinant sodium-iodide symporter. In our study, two hybrid baculovirus systems (Bac-eGFP-SB-NeoR and Bac-NIS-SB-NeoR) were successfully constructed and used to infect U87 glioma cells. After G418 selection screening, the Bac-eGFP-SB-NeoR-U87 cells remained eGFP positive, at the 18th and 196th day post transfection (96.03 ± 0.21% and 97.43 ± 0.81%), while eGFP positive population declined significantly at 18 days in cells transfected with unmodified baculovirus construct. NIS gene expression by Bac-NIS-SB-NeoR-U87 cells was also maintained for 28 weeks as determined by radioiodine uptake assay, reverse transcription-polymerase chain reaction (RT-PCR) and Western Blot (WB) assay. When transplanted in mice, Bac-NIS-SB-NeoR-U87 cells also expressed NIS gene stably as monitored by SPECT imaging for 43 days until the tumor-bearing mice were sacrificed. Herein, we showed that incorporation of SB in Bac-NIS system (hybrid Bac-NIS-SB-NeoR) can achieve a long-term transgene expression and can improve radionuclide imaging in cell tracking and monitoring in vivo.
Collapse
|
22
|
Yoo RJ, Kim MH, Woo SK, Kim KI, Lee TS, Choi YK, Kang JH, Lim SM, Lee YJ. Monitoring of macrophage accumulation in statin-treated atherosclerotic mouse model using sodium iodide symporter imaging system. Nucl Med Biol 2017; 48:45-51. [PMID: 28208058 DOI: 10.1016/j.nucmedbio.2017.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 01/16/2017] [Accepted: 01/20/2017] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Macrophages play a key role in atherosclerotic plaque formation in atherosclerosis, but its detailed understanding has poorly investigated until now. Thus, we sought to demonstrate a noninvasive technique for macrophage tracking to atherosclerotic lesions in apolipoprotein E-/-(ApoE-/-) mice with an imaging system based on sodium iodide symporter (NIS) gene coupled with 99mTc-single-photon emission computed tomography (SPECT). METHODS AND RESULTS Macrophage cells (RAW264.7) were stably transduced with retrovirus expressing NIS gene (RAW-NIS). In RAW-NIS cells, uptake of 125I was higher than the parental cells. [18F]FDG signals in the aorta at 30weeks on an ApoE-/- mice with high cholesterol diet were higher (1.7±0.12% injected dose (ID)) than those in control group (0.84±0.06% ID). Through 99mTc-SPECT/computed tomography (CT), in the RAW-NIS cell injected group, the 99mTc-pertechnetate uptake in aorta was higher than control groups. However, according to atorvastatin treatment, RAW-NIS cell recruitment reduced to the aorta. Area of 99mTc-pertechnetate uptake was positively correlated with immunostaining results against macrophage antigen (CD68). Cholesterol and low-density lipoprotein levels of atorvastatin-treated group showed lower than those of atorvastatin-untreated group, but did not reach statistical difference. CONCLUSIONS This novel approach to tracking macrophages to atherosclerotic plaques in vivo can be applied for studies of arterosclerotic vascular disease.
Collapse
Affiliation(s)
- Ran Ji Yoo
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea; Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Min Hwan Kim
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Sang-Keun Woo
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Kwang Il Kim
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Tae Sup Lee
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Joo Hyun Kang
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Sang Moo Lim
- Department of Nuclear Medicine, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Yong Jin Lee
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Lee SB, Lee HW, Singh TD, Li Y, Kim SK, Cho SJ, Lee SW, Jeong SY, Ahn BC, Choi S, Lee IK, Lim DK, Lee J, Jeon YH. Visualization of Macrophage Recruitment to Inflammation Lesions using Highly Sensitive and Stable Radionuclide-Embedded Gold Nanoparticles as a Nuclear Bio-Imaging Platform. Theranostics 2017; 7:926-934. [PMID: 28382164 PMCID: PMC5381254 DOI: 10.7150/thno.17131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 12/08/2016] [Indexed: 12/20/2022] Open
Abstract
Reliable and sensitive imaging tools are required to track macrophage migration and provide a better understating of their biological roles in various diseases. Here, we demonstrate the possibility of radioactive iodide-embedded gold nanoparticles (RIe-AuNPs) as a cell tracker for nuclear medicine imaging. To demonstrate this utility, we monitored macrophage migration to carrageenan-induced sites of acute inflammation in living subjects and visualized the effects of anti-inflammatory agents on this process. Macrophage labeling with RIe-AuNPs did not alter their biological functions such as cell proliferation, phenotype marker expression, or phagocytic activity. In vivo imaging with positron-emission tomography revealed the migration of labeled macrophages to carrageenan-induced inflammation lesions 3 h after transfer, with highest recruitment at 6 h and a slight decline of radioactive signal at 24 h; these findings were highly consistent with the data of a bio-distribution study. Treatment with dexamethasone (an anti-inflammation drug) or GSK5182 (an ERRγ inverse agonist) hindered macrophage recruitment to the inflamed sites. Our findings suggest that a cell tracking strategy utilizing RIe-AuNPs will likely be highly useful in research related to macrophage-related disease and cell-based therapies.
Collapse
|
24
|
Oh SG, Li X, Lee HW, Singh TD, Lee SB, Ji HD, Yoon G, Cho SJ, Lee IK, Jeong SY, Ahn BC, Lee J, Chang HW, Lee SW, Jeon YH. Non-invasive visualization of mast cell recruitment and its effects in lung cancer by optical reporter gene imaging and glucose metabolism monitoring. Biomaterials 2017; 112:192-203. [DOI: 10.1016/j.biomaterials.2016.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
|
25
|
Abstract
Chimaeric antigen receptor (CAR) therapy is entering the mainstream for the treatment of CD19(+)cancers. As is does we learn more about resistance to therapy and the role, risks and management of toxicity. In solid tumour CAR therapy research the route to the clinic is less smooth with a wealth of challenges facing translating this, potentially hugely valuable, therapeutic option for patients. As we strive to understand our successes, and navigate the challenges, having a clear understanding of how adoptively transferred CAR-T-cells behavein vivoand in human trials is invaluable. Harnessing reporter gene imaging to enable detection and tracking of small numbers of CAR-T-cells after adoptive transfer is one way by which we can accomplish this. The compatibility of certain reporter gene systems with tracers available routinely in the clinic makes this approach highly useful for future appraisal of CAR-T-cell success in humans.
Collapse
|
26
|
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: 29] [Impact Index Per Article: 3.2] [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
|
27
|
Lee HW, Gangadaran P, Kalimuthu S, Ahn BC. Advances in Molecular Imaging Strategies for In Vivo Tracking of Immune Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1946585. [PMID: 27725934 PMCID: PMC5048043 DOI: 10.1155/2016/1946585] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/12/2016] [Accepted: 08/23/2016] [Indexed: 01/25/2023]
Abstract
Tracking of immune cells in vivo is a crucial tool for development and optimization of cell-based therapy. Techniques for tracking immune cells have been applied widely for understanding the intrinsic behavior of immune cells and include non-radiation-based techniques such as optical imaging and magnetic resonance imaging (MRI), radiation-based techniques such as computerized tomography (CT), and nuclear imaging including single photon emission computerized tomography (SPECT) and positron emission tomography (PET). Each modality has its own strengths and limitations. To overcome the limitations of each modality, multimodal imaging techniques involving two or more imaging modalities are actively applied. Multimodal techniques allow integration of the strengths of individual modalities. In this review, we discuss the strengths and limitations of currently available preclinical in vivo immune cell tracking techniques and summarize the value of immune cell tracking in the development and optimization of immune cell therapy for various diseases.
Collapse
Affiliation(s)
- Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, Republic of Korea
| |
Collapse
|
28
|
18F-FBPA as a tumor-specific probe of L-type amino acid transporter 1 (LAT1): a comparison study with 18F-FDG and 11C-Methionine PET. Eur J Nucl Med Mol Imaging 2016; 44:321-331. [PMID: 27550420 DOI: 10.1007/s00259-016-3487-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE The purpose of this study was to evaluate the usefulness of L-4-borono-2-18F-fluoro-phenylalanine (18F-FBPA) as a tumor-specific probe, in comparison to 18F-FDG and 11C-methionine (Met), focusing on its transport selectivity by L-type amino acid transporter 1 (LAT1), which is highly upregulated in cancers. METHODS Cellular analyses of FBPA were performed to evaluate the transportablity and Km value. PET studies were performed in rat xenograft models of C6 glioma (n = 12) and in rat models of turpentine oil-induced subcutaneous inflammation (n = 9). The kinetic parameters and uptake values on static PET images were compared using the one-tissue compartment model (K1, k2) and maximum standardized uptake value (SUVmax). RESULTS The cellular analyses showed that FBPA had a lower affinity to a normal cell-type transporter LAT2 and induced less efflux through LAT2 among FBPA, Met, and BPA, while the efflux through LAT1 induced by FBPA was similar among the three compounds. The Km value of 18F-FBPA for LAT1 (196.8 ± 11.4 μM) was dramatically lower than that for LAT2 (2813.8 ± 574.5 μM), suggesting the higher selectivity of 18F-FBPA for LAT1. K1 and k2 values were significantly smaller in 18F-FBPA PET (K1 = 0.04 ± 0.01 ml/ccm/min and k2 = 0.07 ± 0.01 /min) as compared to 11C-Met PET (0.22 ± 0.09 and 0.52 ± 0.10, respectively) in inflammatory lesions. Static PET analysis based on the SUVmax showed significantly higher accumulation of 18F-FDG in the tumor and inflammatory lesions (7.2 ± 2.1 and 4.6 ± 0.63, respectively) as compared to both 18F-FBPA (3.2 ± 0.40 and 1.9 ± 0.19) and 11C-Met (3.4 ± 0.43 and 1.6 ± 0.11). No significant difference was observed between 18F-FBPA and 11C-Met in the static PET images. CONCLUSION This study shows the utility of 18F-FBPA as a tumor-specific probe of LAT1 with low accumulation in the inflammatory lesions.
Collapse
|
29
|
Kang S, Lee HW, Jeon YH, Singh TD, Choi YJ, Park JY, Kim JS, Lee H, Hong KS, Lee I, Jeong SY, Lee SW, Ha JH, Ahn BC, Lee J. Combined Fluorescence and Magnetic Resonance Imaging of Primary Macrophage Migration to Sites of Acute Inflammation Using Near-Infrared Fluorescent Magnetic Nanoparticles. Mol Imaging Biol 2016; 17:643-51. [PMID: 25669929 DOI: 10.1007/s11307-015-0830-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE This study aimed to track the migration of primary macrophages labeled with near-infrared (NIR) fluorescent magnetic nanoparticles toward chemically induced acute inflammatory lesions in mice and to visualize the effect of anti-inflammatory drugs on macrophage migration using combined fluorescence and magnetic resonance imaging (FLI/MRI). PROCEDURES Primary macrophages were labeled with NIR fluorescent magnetic nanoparticles, and labeled cells were injected into mice intravenously. One day later, inflammation was induced by subcutaneous injection of 1% carrageenan (CG) solution to footpads of the right hind leg, and phosphate-buffered saline (PBS) as control treatment was subcutaneously injected to footpad of the left hind leg. To evaluate the effect of drug treatment on macrophage migration, a single dose of dexamethasone (DEX) was intraperitoneally administered to the mice immediately after the induction of inflammation and was followed by combined FLI/MRI at predetermined time points. RESULTS No difference in cellular viability or phagocytic activity was observed between the labeled and parent macrophages. In vivo optical imaging revealed an increase in FLI signals in CG-injected footpads in a time-dependent manner, but not in PBS-treated footpads. DEX treatment inhibited the migration of the labeled macrophages to the CG-injected footpads, with relative decreases in FLI activity. In accordance with FLI, T2*-weighted MR images showed hypo-intense signals in the CG-injected footpads but not in the PBS-injected footpads. The DEX-treated mice did not show a dark signal loss zone on MR images in the CG-treated paw. CONCLUSIONS We successfully tracked the migration of macrophages to inflammatory lesions using both FLI and MRI with NIR fluorescent magnetic nanoparticles and demonstrated the inhibitory effects of DEX on macrophage migration to inflammation sites.
Collapse
Affiliation(s)
- Sungmin Kang
- Department of Nuclear Medicine, Catholic University of Daegu School of Medicine, Gyeongsan-si, South Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea
| | - Young Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea. .,Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 807 Hogukro, Bukgu, Daegu, 702-210, South Korea.
| | - Thoudam Debraj Singh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea
| | - Yun Ju Choi
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea
| | - Ji Young Park
- Department of Pathology, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Jun Sung Kim
- R&D Center, Biterials, Goyang-si, 410-050, South Korea
| | - Hyunseung Lee
- Division of MR Research, Korea Basic Science Institute, Daejeon, South Korea
| | - Kwan Soo Hong
- Division of MR Research, Korea Basic Science Institute, Daejeon, South Korea
| | - Inkyu Lee
- Department of Endocrinology, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea
| | - Jeoung-Hee Ha
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine, 50 Samduk-dong 2-ga, Chung Gu, Daegu, 700-721, South Korea. .,Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 701-310, Korea.
| |
Collapse
|
30
|
Chung T, Youn H, Yeom CJ, Kang KW, Chung JK. Glycosylation of Sodium/Iodide Symporter (NIS) Regulates Its Membrane Translocation and Radioiodine Uptake. PLoS One 2015; 10:e0142984. [PMID: 26599396 PMCID: PMC4658105 DOI: 10.1371/journal.pone.0142984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/29/2015] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Human sodium/iodide symporter (hNIS) protein is a membrane glycoprotein that transports iodide ions into thyroid cells. The function of this membrane protein is closely regulated by post-translational glycosylation. In this study, we measured glycosylation-mediated changes in subcellular location of hNIS and its function of iodine uptake. METHODS HeLa cells were stably transfected with hNIS/tdTomato fusion gene in order to monitor the expression of hNIS. Cellular localization of hNIS was visualized by confocal microscopy of the red fluorescence of tdTomato. The expression of hNIS was evaluated by RT-PCR and immunoblot analysis. Functional activity of hNIS was estimated by radioiodine uptake. Cyclic AMP (cAMP) and tunicamycin were used to stimulate and inhibit glycosylation, respectively. In vivo images were obtained using a Maestro fluorescence imaging system. RESULTS cAMP-mediated Glycosylation of NIS resulted in increased expression of hNIS, stimulating membrane translocation, and enhanced radioiodine uptake. In contrast, inhibition of glycosylation by treatment with tunicamycin dramatically reduced membrane translocation of intracellular hNIS, resulting in reduced radioiodine uptake. In addition, our hNIS/tdTomato fusion reporter successfully visualized cAMP-induced hNIS expression in xenografted tumors from mouse model. CONCLUSIONS These findings clearly reveal that the membrane localization of hNIS and its function of iodine uptake are glycosylation-dependent, as our results highlight enhancement of NIS expression and glycosylation with subsequent membrane localization after cAMP treatment. Therefore, enhancing functional NIS by the increasing level of glycosylation may be suggested as a promising therapeutic strategy for cancer patients who show refractory response to conventional radioiodine treatment.
Collapse
Affiliation(s)
- Taemoon Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
- * E-mail: (HY); (JKC)
| | - Chan Joo Yeom
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- * E-mail: (HY); (JKC)
| |
Collapse
|
31
|
Micci MA, Boone DR, Parsley MA, Wei J, Patrikeev I, Motamedi M, Hellmich HL. Development of a novel imaging system for cell therapy in the brain. Stem Cell Res Ther 2015; 6:131. [PMID: 26194790 PMCID: PMC4534109 DOI: 10.1186/s13287-015-0129-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/19/2015] [Accepted: 07/09/2015] [Indexed: 01/19/2023] Open
Abstract
Introduction Stem cells have been evaluated as a potential therapeutic approach for several neurological disorders of the central and peripheral nervous system as well as for traumatic brain and spinal cord injury. Currently, the lack of a reliable and safe method to accurately and non-invasively locate the site of implantation and track the migration of stem cells in vivo hampers the development of stem cell therapy and its clinical application. In this report, we present data that demonstrate the feasibility of using the human sodium iodide symporter (hNIS) as a reporter gene for tracking neural stem cells (NSCs) after transplantation in the brain by using single-photon emission tomography/computed tomography (SPECT/CT) imaging. Methods NSCs were isolated from the hippocampus of adult rats (Hipp-NSCs) and transduced with a lentiviral vector containing the hNIS gene. Hipp-NSCs expressing the hNIS (NIS-Hipp-NSCs) were characterized in vitro and in vivo after transplantation in the rat brain and imaged by using technetium-99m (99mTc) and a small rodent SPECT/CT apparatus. Comparisons were made between Hipp-NSCs and NIS-Hipp-NSCs, and statistical analysis was performed by using two-tailed Student’s t test. Results Our results show that the expression of the hNIS allows the repeated visualization of NSCs in vivo in the brain by using SPECT/CT imaging and does not affect the ability of Hipp-NSCs to generate neuronal and glial cells in vitro and in vivo. Conclusions These data support the use of the hNIS as a reporter gene for non-invasive imaging of NSCs in the brain. The repeated, non-invasive tracking of implanted cells will accelerate the development of effective stem cell therapies for traumatic brain injury and other types of central nervous system injury.
Collapse
Affiliation(s)
- Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Debbie R Boone
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Margaret A Parsley
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Jingna Wei
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Igor Patrikeev
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Helen L Hellmich
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
32
|
Lee HW, Yoon SY, Singh TD, Choi YJ, Lee HJ, Park JY, Jeong SY, Lee SW, Ha JH, Ahn BC, Jeon YH, Lee J. Tracking of dendritic cell migration into lymph nodes using molecular imaging with sodium iodide symporter and enhanced firefly luciferase genes. Sci Rep 2015; 5:9865. [PMID: 25974752 PMCID: PMC4431315 DOI: 10.1038/srep09865] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/26/2015] [Indexed: 12/14/2022] Open
Abstract
We sought to evaluate the feasibility of molecular imaging using the human sodium iodide symporter (hNIS) gene as a reporter, in addition to the enhanced firefly luciferase (effluc) gene, for tracking dendritic cell (DCs) migration in living mice. A murine dendritic cell line (DC2.4) co-expressing hNIS and effluc genes (DC/NF) was established. For the DC-tracking study, mice received either parental DCs or DC/NF cells in the left or right footpad, respectively, and combined I-124 PET/CT and bioluminescence imaging (BLI) were performed. In vivo PET/CT imaging with I-124 revealed higher activity of the radiotracer in the draining popliteal lymph nodes (DPLN) of the DC/NF injection site at day 1 than DC injection site (p < 0.05). The uptake value further increased at day 4 (p < 0.005). BLI also demonstrated migration of DC/NF cells to the DPLNs at day 1 post-injection, and signals at the DPLNs were much higher at day 4. These data support the feasibility of hNIS reporter gene imaging in the tracking of DC migration to lymphoid organs in living mice. DCs expressing the NIS reporter gene could be a useful tool to optimize various strategies of cell-based immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Hong Je Lee
- Department of Nuclear Medicine, Dongnam Institution of Radiological &Medical SciencesBusan
| | - Ji Young Park
- Department of Pathology, School of Medicine, Kyungpook National UniversityDaegu
| | | | - Sang-Woo Lee
- 1] Department of Nuclear Medicine [2] Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 807 Hogukro, Bukgu, Daegu
| | | | | | - Yong Hyun Jeon
- 1] Department of Nuclear Medicine [2] Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 807 Hogukro, Bukgu, Daegu
| | - Jaetae Lee
- 1] Department of Nuclear Medicine [2] Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 701-310, Republic of Korea
| |
Collapse
|
33
|
Youniss FM, Sundaresan G, Graham LJ, Wang L, Berry CR, Dewkar GK, Jose P, Bear HD, Zweit J. Near-infrared imaging of adoptive immune cell therapy in breast cancer model using cell membrane labeling. PLoS One 2014; 9:e109162. [PMID: 25334026 PMCID: PMC4204826 DOI: 10.1371/journal.pone.0109162] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/19/2014] [Indexed: 12/22/2022] Open
Abstract
The overall objective of this study is to non-invasively image and assess tumor targeting and retention of directly labeled T-lymphocytes following their adoptive transfer in mice. T-lymphocytes obtained from draining lymph nodes of 4T1 (murine breast cancer cell) sensitized BALB/C mice were activated in-vitro with Bryostatin/Ionomycin for 18 hours, and were grown in the presence of Interleukin-2 for 6 days. T-lymphocytes were then directly labeled with 1,1-dioctadecyltetramethyl indotricarbocyanine Iodide (DiR), a lipophilic near infrared fluorescent dye that labels the cell membrane. Assays for viability, proliferation, and function of labeled T-lymphocytes showed that they were unaffected by DiR labeling. The DiR labeled cells were injected via tail vein in mice bearing 4T1 tumors in the flank. In some cases labeled 4T1 specific T-lymphocytes were injected a week before 4T1 tumor cell implantation. Multi-spectral in vivo fluorescence imaging was done to subtract the autofluorescence and isolate the near infrared signal carried by the T-lymphocytes. In recipient mice with established 4T1 tumors, labeled 4T1 specific T-lymphocytes showed marked tumor retention, which peaked 6 days post infusion and persisted at the tumor site for up to 3 weeks. When 4T1 tumor cells were implanted 1-week post-infusion of labeled T-lymphocytes, T-lymphocytes responded to the immunologic challenge and accumulated at the site of 4T1 cell implantation within two hours and the signal persisted for 2 more weeks. Tumor accumulation of labeled 4T1 specific T-lymphocytes was absent in mice bearing Meth A sarcoma tumors. When lysate of 4T1 specific labeled T-lymphocytes was injected into 4T1 tumor bearing mice the near infrared signal was not detected at the tumor site. In conclusion, our validated results confirm that the near infrared signal detected at the tumor site represents the DiR labeled 4T1 specific viable T-lymphocytes and their response to immunologic challenge can be imaged in vivo.
Collapse
Affiliation(s)
- Fatma M. Youniss
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Gobalakrishnan Sundaresan
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Laura J. Graham
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Li Wang
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Collin R. Berry
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Gajanan K. Dewkar
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Purnima Jose
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Harry D. Bear
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jamal Zweit
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
34
|
Inoue K, Gibbs SL, Liu F, Lee JH, Xie Y, Ashitate Y, Fujii H, Frangioni JV, Choi HS. Microscopic validation of macroscopic in vivo images enabled by same-slide optical and nuclear fusion. J Nucl Med 2014; 55:1899-904. [PMID: 25324521 DOI: 10.2967/jnumed.114.141606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED It is currently difficult to determine the molecular and cellular basis for radioscintigraphic signals obtained during macroscopic in vivo imaging. The field is in need of technology that helps bridge the macroscopic and microscopic regimes. To solve this problem, we developed a fiducial marker (FM) simultaneously compatible with 2-color near-infrared (NIR) fluorescence (700 and 800 nm), autoradiography, and conventional hematoxylin-eosin (HE) histology. METHODS The FM was constructed from an optimized concentration of commercially available human serum albumin, 700- and 800-nm NIR fluorophores, (99m)Tc-pertechnetate, dimethyl sulfoxide, and glutaraldehyde. Lymphangioleiomyomatosis cells coexpressing the sodium iodide symporter and green fluorescent protein were labeled with 700-nm fluorophore and (99m)Tc-pertechnatate and then administered intratracheally into CD-1 mice. After in vivo SPECT imaging and ex vivo SPECT and NIR fluorescence imaging of the lungs, 30-μm frozen sections were prepared and processed for 800-nm NIR fluorophore costaining, autoradiography, and HE staining on the same slide using the FMs to coregister all datasets. RESULTS Optimized FMs, composed of 100 μM unlabeled human serum albumin, 1 μM NIR fluorescent human serum albumin, 15% dimethyl sulfoxide, and 3% glutaraldehyde in phosphate-buffered saline (pH 7.4), were prepared within 15 min, displayed homogeneity and stability, and were visible by all imaging modalities, including HE staining. Using these FMs, tissue displaying high signal by SPECT could be dissected and analyzed on the same slide and at the microscopic level for 700-nm NIR fluorescence, 800-nm NIR fluorescence, autoradiography, and HE histopathologic staining. CONCLUSION When multimodal FMs are combined with a new technique for simultaneous same-slide NIR fluorescence imaging, autoradiography, and HE staining, macroscopic in vivo images can now be studied unambiguously at the microscopic level.
Collapse
Affiliation(s)
- Kazumasa Inoue
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts Department of Radiological Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Summer L Gibbs
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Fangbing Liu
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Jeong Heon Lee
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Yang Xie
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Yoshitomo Ashitate
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Hirofumi Fujii
- Functional Imaging Division, Research Center for Innovative Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - John V Frangioni
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts; and Curadel, LLC, Worcester, Massachusetts
| | - Hak Soo Choi
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Lee HW, Jeon YH, Hwang MH, Kim JE, Park TI, Ha JH, Lee SW, Ahn BC, Lee J. Dual reporter gene imaging for tracking macrophage migration using the human sodium iodide symporter and an enhanced firefly luciferase in a murine inflammation model. Mol Imaging Biol 2014; 15:703-12. [PMID: 23677652 DOI: 10.1007/s11307-013-0645-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE The purpose of this study is to visualize the migration of reporter macrophages expressing both the human sodium iodide symporter (hNIS) and enhanced firefly luciferase (effluc) gene in mice with chemically induced inflammation. PROCEDURES A macrophage cell line expressing both hNIS and effluc genes (Raw264.7/hNIS-effluc, herein referred to as a Raw264.7/NF) was established by cotransduction of two genes into a murine macrophage cell line (Raw264.7), and cell proliferation and phagocytic activity were compared between parental Raw264.7 and Raw264.7/NF cells. Both serial bioluminescence imaging (BLI) and small animal positron emission tomography (PET) imaging with I-124 were performed in inflammation-induced mice at various time points after intravenous injection of either Raw264.7 or Raw264.7/NF cells. RESULTS There was no significant difference in cellular proliferation and phagocytic activity between parental Raw264.7 and Raw264.7/NF cells. Early distribution of Raw264.7/NF cells was successfully visualized in the lung and spleen by BLI, but not by I-124 PET imaging. BLI signals, but not PET signals, were observed from the inflammation site at day 4 after the injection of Raw264.7/NF cells, and the signal intensity gradually increased until day 8. In contrast, focal uptake of I-124 was first detected at the site of inflammation at postinjection day 8, and signal intensity from the inflamed lesion was highest at that time point. While visualization of the inflamed lesion was possible by both BLI and PET imaging until day 14, it was only possible by BLI until day 21 after injection. CONCLUSIONS Tracking of macrophage migration toward inflammation foci was successfully achieved in vivo from early time points by dual reporter gene imaging with a combination of nuclear and optical reporters. Multimodal reporter imaging of macrophages might successfully overcome the limitations of single reporter gene imaging in preclinical models of inflammation.
Collapse
Affiliation(s)
- Ho Won Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, 50 Samduk 2-ga, Daegu, 700-721, Republic of Korea,
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fruhwirth GO, Diocou S, Blower PJ, Ng T, Mullen G. A whole-body dual-modality radionuclide optical strategy for preclinical imaging of metastasis and heterogeneous treatment response in different microenvironments. J Nucl Med 2014; 55:686-94. [PMID: 24604910 PMCID: PMC6205625 DOI: 10.2967/jnumed.113.127480] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Imaging spontaneous cancer cell metastasis or heterogeneous tumor responses to drug treatment in vivo is difficult to achieve. The goal was to develop a new highly sensitive and reliable preclinical longitudinal in vivo imaging model for this purpose, thereby facilitating discovery and validation of anticancer therapies or molecular imaging agents. METHODS The strategy is based on breast cancer cells stably expressing the human sodium iodide symporter (NIS) fused to a red fluorescent protein, thereby permitting radionuclide and fluorescence imaging. Using whole-body nano-SPECT/CT with (99m)TcO4(-), we followed primary tumor growth and spontaneous metastasis in the presence or absence of etoposide treatment. NIS imaging was used to classify organs as small as individual lymph nodes (LNs) to be positive or negative for metastasis, and results were confirmed by confocal fluorescence microscopy. Etoposide treatment efficacy was proven by ex vivo anticaspase 3 staining and fluorescence microscopy. RESULTS In this preclinical model, we found that the NIS imaging strategy outperformed state-of-the-art (18)F-FDG imaging in its ability to detect small tumors (18.5-fold-better tumor-to-blood ratio) and metastases (LN, 3.6-fold) because of improved contrast in organs close to metastatic sites (12- and 8.5-fold-lower standardized uptake value in the heart and kidney, respectively). We applied the model to assess the treatment response to the neoadjuvant etoposide and found a consistent and reliable improvement in spontaneous metastasis detection. Importantly, we also found that tumor cells in different microenvironments responded in a heterogeneous manner to etoposide treatment, which could be determined only by the NIS-based strategy and not by (18)F-FDG imaging. CONCLUSION We developed a new strategy for preclinical longitudinal in vivo cancer cell tracking with greater sensitivity and reliability than (18)F-FDG PET and applied it to track spontaneous and distant metastasis in the presence or absence of genotoxic stress therapy. Importantly, the model provides sufficient sensitivity and dynamic range to permit the reliable assessment of heterogeneous treatment responses in various microenvironments.
Collapse
Affiliation(s)
- Gilbert O. Fruhwirth
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- The Richard Dimbleby Department of Cancer Research, KCL, London SE1 1UL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Seckou Diocou
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Philip J. Blower
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Tony Ng
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- The Richard Dimbleby Department of Cancer Research, KCL, London SE1 1UL
| | - Greg Mullen
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| |
Collapse
|
37
|
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: 192] [Impact Index Per Article: 17.5] [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
|
38
|
Ahn BC. Requisites for successful theranostics with radionuclide-based reporter gene imaging. J Drug Target 2014; 22:295-303. [PMID: 24417717 DOI: 10.3109/1061186x.2013.878940] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radionuclide-based theranostic strategy has been widely used in diagnosis and treatment of patients with hyperthyroidism or differentiated thyroid cancer for a long time, and sodium iodide symporter gene is the radionuclide-based reporter gene used in theranostics. Theranostics, which is a promising approach, offering the ideal combination of accurate diagnosis and successful therapy in various clinical fields, is expected to become a key area of personalized medicine. Rapid advancements in biotechnologies using theranostic reporter genes and theranostic radiochemistry have led to development of the concept of theranostics using radionuclide-based imaging reporter genes; the theranostic approach is almost ready for application in a limited arena of clinics. In order to fulfill both the diagnostic and therapeutic purposes, theranostics with radionuclide-based imaging reporter requires use of successful combinations of various components, such as radionuclide-based reporter genes, promoters/enhancers that regulate expression of reporter genes, delivery vectors/vehicles, imaging or therapeutic probes and prodrugs, transductional and transcriptional targeting strategies, transgene amplification systems, etc. In this review, overview and recent updates on theranostics using radionuclide-based imaging reporter genes will be discussed.
Collapse
Affiliation(s)
- Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital , Daegu , Republic of Korea
| |
Collapse
|
39
|
Nawroth I, Alsner J, Deleuran BW, Dagnaes-Hansen F, Yang C, Horsman MR, Overgaard J, Howard KA, Kjems J, Gao S. Peritoneal macrophages mediated delivery of chitosan/siRNA nanoparticle to the lesion site in a murine radiation-induced fibrosis model. Acta Oncol 2013; 52:1730-8. [PMID: 23020526 DOI: 10.3109/0284186x.2012.726373] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Radiation-induced fibrosis (RIF) is a dose-limiting complication of cancer radiotherapy and causes serious problems, i.e. restricted tissue flexibility, pain, ulceration or necrosis. Recently, we have successfully treated RIF in a mouse model by intraperitoneal administration of chitosan/siRNA nanoparticles directed towards silencing TNF alpha in local macrophage populations, but the mechanism for the therapeutic effect at the lesion site remains unclear. METHODS Using the same murine RIF model we utilized an optical imaging technique and fluorescence microscopy to investigate the uptake of chitosan/fluorescently labeled siRNA nanoparticles by peritoneal macrophages and their subsequent migration to the inflamed tissue in the RIF model. RESULTS We observed strong accumulation of the fluorescent signal in the lesion site of the irradiated leg up to 24 hours using the optical imaging system. We further confirm by immunohistochemical staining that Cy3 labeled siRNA resides in macrophages of the irradiated leg. CONCLUSION We provide a proof-of-concept for host macrophage trafficking towards the inflamed region in a murine RIF model, which thereby suggests that the chitosan/siRNA nanoparticle may constitute a general treatment for inflammatory diseases using the natural homing potential of macrophages to inflammatory sites.
Collapse
Affiliation(s)
- Isabel Nawroth
- Interdisciplinary Nanoscience Center (iNANO) , Aarhus University, Aarhus C , Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Penheiter AR, Russell SJ, Carlson SK. The sodium iodide symporter (NIS) as an imaging reporter for gene, viral, and cell-based therapies. Curr Gene Ther 2012; 12:33-47. [PMID: 22263922 PMCID: PMC3367315 DOI: 10.2174/156652312799789235] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 01/04/2012] [Accepted: 01/06/2012] [Indexed: 02/06/2023]
Abstract
Preclinical and clinical tomographic imaging systems increasingly are being utilized for non-invasive imaging of reporter gene products to reveal the distribution of molecular therapeutics within living subjects. Reporter gene and probe combinations can be employed to monitor vectors for gene, viral, and cell-based therapies. There are several reporter systems available; however, those employing radionuclides for positron emission tomography (PET) or singlephoton emission computed tomography (SPECT) offer the highest sensitivity and the greatest promise for deep tissue imaging in humans. Within the category of radionuclide reporters, the thyroidal sodium iodide symporter (NIS) has emerged as one of the most promising for preclinical and translational research. NIS has been incorporated into a remarkable variety of viral and non-viral vectors in which its functionality is conveniently determined by in vitro iodide uptake assays prior to live animal imaging. This review on the NIS reporter will focus on 1) differences between endogenous NIS and heterologously-expressed NIS, 2) qualitative or comparative use of NIS as an imaging reporter in preclinical and translational gene therapy, oncolytic viral therapy, and cell trafficking research, and 3) use of NIS as an absolute quantitative reporter.
Collapse
Affiliation(s)
- Alan R Penheiter
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | |
Collapse
|
41
|
Ahn BC. Sodium iodide symporter for nuclear molecular imaging and gene therapy: from bedside to bench and back. Theranostics 2012; 2:392-402. [PMID: 22539935 PMCID: PMC3337731 DOI: 10.7150/thno.3722] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/23/2012] [Indexed: 12/20/2022] Open
Abstract
Molecular imaging, defined as the visual representation, characterization and quantification of biological processes at the cellular and subcellular levels within intact living organisms, can be obtained by various imaging technologies, including nuclear imaging methods. Imaging of normal thyroid tissue and differentiated thyroid cancer, and treatment of thyroid cancer with radioiodine rely on the expression of the sodium iodide symporter (NIS) in these cells. NIS is an intrinsic membrane protein with 13 transmembrane domains and it takes up iodide into the cytosol from the extracellular fluid. By transferring NIS function to various cells via gene transfer, the cells can be visualized with gamma or positron emitting radioisotopes such as Tc-99m, I-123, I-131, I-124 and F-18 tetrafluoroborate, which are accumulated by NIS. They can also be treated with beta- or alpha-emitting radionuclides, such as I-131, Re-186, Re-188 and At-211, which are also accumulated by NIS. This article demonstrates the diagnostic and therapeutic applications of NIS as a radionuclide-based reporter gene for trafficking cells and a therapeutic gene for treating cancers.
Collapse
Affiliation(s)
- Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, Daegu, South Korea
| |
Collapse
|
42
|
Graham MM. Clinical molecular imaging with radiotracers: current status. Med Princ Pract 2012; 21:197-208. [PMID: 22142905 DOI: 10.1159/000333552] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 09/05/2011] [Indexed: 12/17/2022] Open
Abstract
Molecular imaging is defined as the visualization, characterization, and measurement of biological processes at the molecular and cellular levels in humans and other living systems. Most clinical molecular imaging is currently done using radioisotope-labeled agents to define the activity of various metabolic pathways in vivo or to determine the distribution and density of various receptors relevant to human disease. This paper briefly reviews most of the commonly used radiopharmaceuticals in nuclear medicine, as well as newer agents that are likely to become available in the near future. The metabolic pathways include those relevant to the thyroid, parathyroid, heart, brain, bones, kidneys, liver, pancreas, adrenals and tumor. The receptor systems include agents useful in evaluating movement disorders, dementia, cardiac sympathetic enervation and neoangiogenesis. Receptor systems relevant to tumors include somatostatin receptors (neuroendocrine tumors), prostate-specific membrane antigen, carbonic anhydrase IX (renal cancer), and CD-20 (lymphoma). These agents, and newer agents that are being developed, are likely to become critical in the development of personalized medicine, where it will become increasingly important to determine whether a treatment that is targeted to a specific metabolic pathway or receptor is likely to be successful.
Collapse
Affiliation(s)
- Michael M Graham
- Department of Radiology, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|