1
|
Baena JC, Pérez LM, Toro-Pedroza A, Kitawaki T, Loukanov A. CAR T Cell Nanosymbionts: Revealing the Boundless Potential of a New Dyad. Int J Mol Sci 2024; 25:13157. [PMID: 39684867 DOI: 10.3390/ijms252313157] [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: 09/01/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer treatment has traditionally focused on eliminating tumor cells but faces challenges such as resistance and toxicity. A promising direction involves targeting the tumor microenvironment using CAR T cell immunotherapy, which has shown potential for treating relapsed and refractory cancers but is limited by high costs, resistance, and toxicity, especially in solid tumors. The integration of nanotechnology into ICAM cell therapy, a concept we have named "CAR T nanosymbiosis", offers new opportunities to overcome these challenges. Nanomaterials can enhance CAR T cell delivery, manufacturing, activity modulation, and targeting of the tumor microenvironment, providing better control and precision. This approach aims to improve the efficacy of CAR T cells against solid tumors, reduce associated toxicities, and ultimately enhance patient outcomes. Several studies have shown promising results, and developing this therapy further is essential for increasing its accessibility and effectiveness. Our "addition by subtraction model" synthesizes these multifaceted elements into a unified strategy to advance cancer treatment paradigms.
Collapse
Affiliation(s)
- Juan C Baena
- Division of Oncology, Department of Medicine, Fundación Valle del Lili, ICESI University, Carrera 98 No. 18-49, Cali 760032, Colombia
- LiliCAR-T Group, Fundación Valle del Lili, ICESI University, Cali 760032, Colombia
| | - Lucy M Pérez
- Division of Oncology, Department of Medicine, Fundación Valle del Lili, ICESI University, Carrera 98 No. 18-49, Cali 760032, Colombia
- LiliCAR-T Group, Fundación Valle del Lili, ICESI University, Cali 760032, Colombia
| | - Alejandro Toro-Pedroza
- Division of Oncology, Department of Medicine, Fundación Valle del Lili, ICESI University, Carrera 98 No. 18-49, Cali 760032, Colombia
- LiliCAR-T Group, Fundación Valle del Lili, ICESI University, Cali 760032, Colombia
| | - Toshio Kitawaki
- Department of Hematology, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Alexandre Loukanov
- Department of Chemistry and Materials Science, National Institute of Technology, Gunma College, Maebashi 371-8530, Japan
- Laboratory of Engineering Nanobiotechnology, University of Mining and Geology "St. Ivan Rilski", 1700 Sofia, Bulgaria
| |
Collapse
|
2
|
Zeng Q, Liu Z, Niu T, He C, Qu Y, Qian Z. Application of nanotechnology in CAR-T-cell immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
3
|
Evaluation of [18F]tetrafluoroborate as a Potential PET Imaging Agent in a Sodium Iodide Symporter-Transfected Cell Line A549 and Endogenous NIS-Expressing Cell Lines MKN45 and K1. Mol Imaging 2022; 2022:2679260. [PMID: 35330799 PMCID: PMC8923191 DOI: 10.1155/2022/2679260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
[18F]tetrafluoroborate (TFB) has been introduced as the 18F-labeled PET imaging probe for the human sodium iodide symporter (NIS). Noninvasive NIS imaging using [18F]TFB has received much interest in recent years for evaluating various NIS-expressing tumors. Cancers are a global concern with enormous implications; therefore, improving diagnostic methods for accurate detection of cancer is extremely important. Our aim was to investigate the PET imaging capabilities of [18F]TFB in NIS-transfected lung cell line A549 and endogenous NIS-expressing tumor cells, such as thyroid cancer K1 and gastric cancer MKN45, and broaden its application in the medical field. Western blot and flow cytometry were used to assess the NIS expression level. Radioactivity counts of [18F]TFB, in vitro, in the three tumor cells were substantially higher than those in the KI inhibition group in the uptake experiment. In vivo PET imaging clearly delineated the three tumors based on the specific accumulation of [18F]TFB in a mouse model. Ex vivo biodistribution investigation showed high [18F]TFB absorption in the tumor location, which was consistent with the PET imaging results. These results support the use of NIS-transfected lung cell line A549 and NIS-expressing tumor cells MKN45 and K1, to investigate probing capabilities of [18F]TFB. We also demonstrate, for the first time, the feasibility of [18F]TFB in diagnosing stomach cancer. In conclusion, this study illustrates the promising future of [18F]TFB for tumor diagnosis and NIS reporter imaging.
Collapse
|
4
|
Islam A, Pishesha N, Harmand TJ, Heston H, Woodham AW, Cheloha RW, Bousbaine D, Rashidian M, Ploegh HL. Converting an Anti-Mouse CD4 Monoclonal Antibody into an scFv Positron Emission Tomography Imaging Agent for Longitudinal Monitoring of CD4 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1468-1477. [PMID: 34408009 PMCID: PMC8387391 DOI: 10.4049/jimmunol.2100274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
Immuno-positron emission tomography (PET), a noninvasive imaging modality, can provide a dynamic approach for longitudinal assessment of cell populations of interest. Transformation of mAbs into single-chain variable fragment (scFv)-based PET imaging agents would allow noninvasive tracking in vivo of a wide range of possible targets. We used sortase-mediated enzymatic labeling in combination with PEGylation to develop an anti-mouse CD4 scFv-based PET imaging agent constructed from an anti-mouse CD4 mAb. This anti-CD4 scFv can monitor the in vivo distribution of CD4+ T cells by immuno-PET. We tracked CD4+ and CD8+ T cells in wild-type mice, in immunodeficient recipients reconstituted with monoclonal populations of OT-II and OT-I T cells, and in a B16 melanoma model. Anti-CD4 and -CD8 immuno-PET showed that the persistence of both CD4+ and CD8+ T cells transferred into immunodeficient mice improved when recipients were immunized with OVA in CFA. In tumor-bearing animals, infiltration of both CD4+ and CD8+ T cells increased as the tumor grew. The approach described in this study should be readily applicable to convert clinically useful Abs into the corresponding scFv PET imaging agents.
Collapse
Affiliation(s)
- Ashraful Islam
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Society of Fellows, Harvard University, Cambridge, MA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Thibault J Harmand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Hailey Heston
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Djenet Bousbaine
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA; and
- Department of Radiology, Harvard Medical School, Boston, MA
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA;
- Department of Pediatrics, Harvard Medical School, Boston, MA
| |
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
|
Xie T, Chen X, Fang J, Xue W, Zhang J, Tong H, Liu H, Guo Y, Yang Y, Zhang W. Non-invasive monitoring of the kinetic infiltration and therapeutic efficacy of nanoparticle-labeled chimeric antigen receptor T cells in glioblastoma via 7.0-Tesla magnetic resonance imaging. Cytotherapy 2020; 23:211-222. [PMID: 33334686 DOI: 10.1016/j.jcyt.2020.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND AIMS Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment strategy in solid tumors. In vivo cell tracking techniques can help us better understand the infiltration, persistence and therapeutic efficacy of CAR T cells. In this field, magnetic resonance imaging (MRI) can achieve high-resolution images of cells by using cellular imaging probes. MRI can also provide various biological information on solid tumors. METHODS The authors adopted the amino alcohol derivatives of glucose-coated nanoparticles, ultra-small superparamagnetic particles of iron oxide (USPIOs), to label CAR T cells for non-invasive monitoring of kinetic infiltration and persistence in glioblastoma (GBM). The specific targeting CARs included anti-human epidermal growth factor receptor variant III and IL13 receptor subunit alpha 2 CARs. RESULTS When using an appropriate concentration, USPIO labeling exerted no negative effects on the biological characteristics and killing efficiency of CAR T cells. Increasing hypointensity signals could be detected in GBM models by susceptibility-weighted imaging MRI ranging from 3 days to 14 days following the injection of USPIO-labeled CAR T cells. In addition, nanoparticles and CAR T cells were found on consecutive histopathological sections. Moreover, diffusion and perfusion MRI revealed significantly increased water diffusion and decreased vascular permeability on day 3 after treatment, which was simultaneously accompanied by a significant decrease in tumor cell proliferation and increase in intercellular tight junction on immunostaining sections. CONCLUSION These results establish an effective imaging technique that can track CAR T cells in GBM models and validate their early therapeutic effects, which may guide the evaluation of CAR T-cell therapies in solid tumors.
Collapse
Affiliation(s)
- Tian Xie
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Jingqin Fang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Wei Xue
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Junfeng Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yu Guo
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China
| | - Yizeng Yang
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China; Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing, China.
| |
Collapse
|
7
|
Werner JM, Lohmann P, Fink GR, Langen KJ, Galldiks N. Current Landscape and Emerging Fields of PET Imaging in Patients with Brain Tumors. Molecules 2020; 25:E1471. [PMID: 32213992 PMCID: PMC7146177 DOI: 10.3390/molecules25061471] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The number of positron-emission tomography (PET) tracers used to evaluate patients with brain tumors has increased substantially over the last years. For the management of patients with brain tumors, the most important indications are the delineation of tumor extent (e.g., for planning of resection or radiotherapy), the assessment of treatment response to systemic treatment options such as alkylating chemotherapy, and the differentiation of treatment-related changes (e.g., pseudoprogression or radiation necrosis) from tumor progression. Furthermore, newer PET imaging approaches aim to address the need for noninvasive assessment of tumoral immune cell infiltration and response to immunotherapies (e.g., T-cell imaging). This review summarizes the clinical value of the landscape of tracers that have been used in recent years for the above-mentioned indications and also provides an overview of promising newer tracers for this group of patients.
Collapse
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Gereon R. Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
- Department of Nuclear Medicine, University Hospital Aachen, 52074 Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| |
Collapse
|
8
|
Wei W, Jiang D, Ehlerding EB, Luo Q, Cai W. Noninvasive PET Imaging of T cells. Trends Cancer 2018; 4:359-373. [PMID: 29709260 DOI: 10.1016/j.trecan.2018.03.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 02/07/2023]
Abstract
The rapidly evolving field of cancer immunotherapy recently saw the approval of several new therapeutic antibodies. Several cell therapies, for example, chimeric antigen receptor-expressing T cells (CAR-T), are currently in clinical trials for a variety of cancers and other diseases. However, approaches to monitor changes in the immune status of tumors or to predict therapeutic responses are limited. Monitoring lymphocytes from whole blood or biopsies does not provide dynamic and spatial information about T cells in heterogeneous tumors. Positron emission tomography (PET) imaging using probes specific for T cells can noninvasively monitor systemic and intratumoral immune alterations during experimental therapies and may have an important and expanding value in the clinic.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Department of Radiology, Department of Medical Physics, University of Wisconsin, Madison, WI 53705, USA; These authors contributed equally to this work
| | - Dawei Jiang
- Department of Radiology, Department of Medical Physics, University of Wisconsin, Madison, WI 53705, USA; These authors contributed equally to this work
| | - Emily B Ehlerding
- Department of Medical Physics, University of Wisconsin, Madison, WI 53705, USA
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Weibo Cai
- Department of Radiology, Department of Medical Physics, University of Wisconsin, Madison, WI 53705, USA; Department of Medical Physics, University of Wisconsin, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, USA.
| |
Collapse
|
9
|
Thunemann M, Schörg BF, Feil S, Lin Y, Voelkl J, Golla M, Vachaviolos A, Kohlhofer U, Quintanilla-Martinez L, Olbrich M, Ehrlichmann W, Reischl G, Griessinger CM, Langer HF, Gawaz M, Lang F, Schäfers M, Kneilling M, Pichler BJ, Feil R. Cre/lox-assisted non-invasive in vivo tracking of specific cell populations by positron emission tomography. Nat Commun 2017; 8:444. [PMID: 28874662 PMCID: PMC5585248 DOI: 10.1038/s41467-017-00482-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 07/03/2017] [Indexed: 01/15/2023] Open
Abstract
Many pathophysiological processes are associated with proliferation, migration or death of distinct cell populations. Monitoring specific cell types and their progeny in a non-invasive, longitudinal and quantitative manner is still challenging. Here we show a novel cell-tracking system that combines Cre/lox-assisted cell fate mapping with a thymidine kinase (sr39tk) reporter gene for cell detection by positron emission tomography (PET). We generate Rosa26-mT/sr39tk PET reporter mice and induce sr39tk expression in platelets, T lymphocytes or cardiomyocytes. As proof of concept, we demonstrate that our mouse model permits longitudinal PET imaging and quantification of T-cell homing during inflammation and cardiomyocyte viability after myocardial infarction. Moreover, Rosa26-mT/sr39tk mice are useful for whole-body characterization of transgenic Cre mice and to detect previously unknown Cre activity. We anticipate that the Cre-switchable PET reporter mice will be broadly applicable for non-invasive long-term tracking of selected cell populations in vivo.Non-invasive cell tracking is a powerful method to visualize cells in vivo under physiological and pathophysiological conditions. Here Thunemann et al. generate a mouse model for in vivo tracking and quantification of specific cell types by combining a PET reporter gene with Cre-dependent activation that can be exploited for any cell population for which a Cre mouse line is available.
Collapse
Affiliation(s)
- Martin Thunemann
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany.,Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Barbara F Schörg
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany
| | - Yun Lin
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Jakob Voelkl
- Physiologisches Institut I, University of Tübingen, 72076 Tübingen, Germany
| | - Matthias Golla
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany
| | - Angelos Vachaviolos
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany
| | - Ursula Kohlhofer
- Institute of Pathology and Neuropathology, University of Tübingen, and Comprehensive Cancer Center, University Hospital, 72076 Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, University of Tübingen, and Comprehensive Cancer Center, University Hospital, 72076 Tübingen, Germany
| | - Marcus Olbrich
- Department of Cardiovascular Medicine, University Hospital, University of Tübingen, 72076 Tübingen, Germany
| | - Walter Ehrlichmann
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Christoph M Griessinger
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Harald F Langer
- Department of Cardiovascular Medicine, University Hospital, University of Tübingen, 72076 Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiovascular Medicine, University Hospital, University of Tübingen, 72076 Tübingen, Germany
| | - Florian Lang
- Physiologisches Institut I, University of Tübingen, 72076 Tübingen, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital, European Institute for Molecular Imaging & EXC 1003 Cells-in-Motion Cluster of Excellence, University of Münster, 48149 Münster, Germany
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany.,Department of Dermatology, University Hospital, University of Tübingen, 72076 Tübingen, Germany
| | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
10
|
Freise AC, Wu AM. In vivo imaging with antibodies and engineered fragments. Mol Immunol 2015; 67:142-52. [PMID: 25934435 PMCID: PMC4529772 DOI: 10.1016/j.molimm.2015.04.001] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Antibodies have clearly demonstrated their utility as therapeutics, providing highly selective and effective drugs to treat diseases in oncology, hematology, cardiology, immunology and autoimmunity, and infectious diseases. More recently, a pressing need for equally specific and targeted imaging agents for assessing disease in vivo, in preclinical models and patients, has emerged. This review summarizes strategies for developing and optimizing antibodies as targeted probes for use in non-invasive imaging using radioactive, optical, magnetic resonance, and ultrasound approaches. Recent advances in engineered antibody fragments and scaffolds, conjugation and labeling methods, and multimodality probes are highlighted. Importantly, antibody-based imaging probes are seeing new applications in detection and quantitation of cell surface biomarkers, imaging specific responses to targeted therapies, and monitoring immune responses in oncology and other diseases. Antibody-based imaging will provide essential tools to facilitate the transition to truly precision medicine.
Collapse
Affiliation(s)
- Amanda C Freise
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, USA
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, USA.
| |
Collapse
|
11
|
McCracken MN, Vatakis DN, Dixit D, McLaughlin J, Zack JA, Witte ON. Noninvasive detection of tumor-infiltrating T cells by PET reporter imaging. J Clin Invest 2015; 125:1815-26. [PMID: 25822024 DOI: 10.1172/jci77326] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 02/19/2015] [Indexed: 11/17/2022] Open
Abstract
Adoptive transfer of tumor-reactive T cells can successfully reduce tumor burden; however, in rare cases, lethal on-target/off-tumor effects have been reported. A noninvasive method to track engineered cells with high sensitivity and resolution would allow observation of correct cell homing and/or identification of dangerous off-target locations in preclinical and clinical applications. Human deoxycytidine kinase triple mutant (hdCK3mut) is a nonimmunogenic PET reporter that was previously shown to be an effective tool to monitor whole-body hematopoiesis. Here, we engineered a construct in which hdCK3mut is coexpressed with the anti-melanoma T cell receptor F5, introduced this construct into human CD34 cells or PBMCs, and evaluated this approach in multiple immunotherapy models. Expression of hdCK3mut allowed engrafted cells to be visualized within recipient bone marrow, while accumulation of [18F]-L-FMAU in hdCK3mut-expressing T cells permitted detection of intratumoral homing. Animals that received T cells coexpressing hdCK3mut and the anti-melanoma T cell receptor had demonstrably higher signals in HLA-matched tumors compared with those in animals that received cells solely expressing hdCK3mut. Engineered T cells caused cytotoxicity in HLA/antigen-matched tumors and induced IFN-γ production and activation. Moreover, hdCK3mut permitted simultaneous monitoring of engraftment and tumor infiltration, without affecting T cell function. Our findings suggest that hdCK3mut reporter imaging can be applied in clinical immunotherapies for whole-body detection of engineered cell locations.
Collapse
MESH Headings
- Animals
- Bone Marrow/diagnostic imaging
- Chemotaxis, Leukocyte
- Cytotoxicity Tests, Immunologic
- Deoxycytidine Kinase/analysis
- Deoxycytidine Kinase/genetics
- Genes, Reporter
- Genes, Synthetic
- Genetic Vectors/genetics
- Graft Survival
- HLA-A2 Antigen/immunology
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/chemistry
- Hematopoietic Stem Cells/physiology
- Humans
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Interferon-gamma Release Tests
- Lentivirus/genetics
- Leukocytes, Mononuclear/chemistry
- Leukocytes, Mononuclear/physiology
- Leukocytes, Mononuclear/transplantation
- Lymphocytes, Tumor-Infiltrating/immunology
- MART-1 Antigen/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mutation
- Positron-Emission Tomography
- Receptors, Antigen, T-Cell/analysis
- Receptors, Antigen, T-Cell/genetics
- Recombinant Fusion Proteins/analysis
- Recombinant Fusion Proteins/genetics
- Retroviridae/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Thymus Gland/transplantation
Collapse
|
12
|
Quintela BDM, dos Santos RW, Lobosco M. On the coupling of two models of the human immune response to an antigen. BIOMED RESEARCH INTERNATIONAL 2014; 2014:410457. [PMID: 25140313 PMCID: PMC4130187 DOI: 10.1155/2014/410457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/15/2014] [Accepted: 04/15/2014] [Indexed: 12/24/2022]
Abstract
The development of mathematical models of the immune response allows a better understanding of the multifaceted mechanisms of the defense system. The main purpose of this work is to present a scheme for coupling distinct models of different scales and aspects of the immune system. As an example, we propose a new model where the local tissue inflammation processes are simulated with partial differential equations (PDEs) whereas a system of ordinary differential equations (ODEs) is used as a model for the systemic response. The simulation of distinct scenarios allows the analysis of the dynamics of various immune cells in the presence of an antigen. Preliminary results of this approach with a sensitivity analysis of the coupled model are shown but further validation is still required.
Collapse
Affiliation(s)
- Bárbara de M. Quintela
- Laboratory of Computational Physiology and High-Performance Computing (FISIOCOMP), Graduate Program in Computational Modeling, UFJF, Rua José Lourenço Kelmer s/n, Campus Universitário, Bairro São Pedro, 36036-900 Juiz de Fora, MG, Brazil
| | - Rodrigo Weber dos Santos
- Laboratory of Computational Physiology and High-Performance Computing (FISIOCOMP), Graduate Program in Computational Modeling, UFJF, Rua José Lourenço Kelmer s/n, Campus Universitário, Bairro São Pedro, 36036-900 Juiz de Fora, MG, Brazil
| | - Marcelo Lobosco
- Laboratory of Computational Physiology and High-Performance Computing (FISIOCOMP), Graduate Program in Computational Modeling, UFJF, Rua José Lourenço Kelmer s/n, Campus Universitário, Bairro São Pedro, 36036-900 Juiz de Fora, MG, Brazil
| |
Collapse
|
13
|
Tumor lysing genetically engineered T cells loaded with multi-modal imaging agents. Sci Rep 2014; 4:4502. [PMID: 24675806 PMCID: PMC3968458 DOI: 10.1038/srep04502] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/04/2014] [Indexed: 01/11/2023] Open
Abstract
Genetically-modified T cells expressing chimeric antigen receptors (CAR) exert anti-tumor effect by identifying tumor-associated antigen (TAA), independent of major histocompatibility complex. For maximal efficacy and safety of adoptively transferred cells, imaging their biodistribution is critical. This will determine if cells home to the tumor and assist in moderating cell dose. Here, T cells are modified to express CAR. An efficient, non-toxic process with potential for cGMP compliance is developed for loading high cell number with multi-modal (PET-MRI) contrast agents (Super Paramagnetic Iron Oxide Nanoparticles - Copper-64; SPION-(64)Cu). This can now be potentially used for (64)Cu-based whole-body PET to detect T cell accumulation region with high-sensitivity, followed by SPION-based MRI of these regions for high-resolution anatomically correlated images of T cells. CD19-specific-CAR(+)SPION(pos) T cells effectively target in vitro CD19(+) lymphoma.
Collapse
|
14
|
Griessinger CM, Kehlbach R, Bukala D, Wiehr S, Bantleon R, Cay F, Schmid A, Braumüller H, Fehrenbacher B, Schaller M, Eichner M, Sutcliffe JL, Ehrlichmann W, Eibl O, Reischl G, Cherry SR, Röcken M, Pichler BJ, Kneilling M. In Vivo Tracking of Th1 Cells by PET Reveals Quantitative and Temporal Distribution and Specific Homing in Lymphatic Tissue. J Nucl Med 2014; 55:301-7. [DOI: 10.2967/jnumed.113.126318] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
15
|
Engineered antibody fragments for immuno-PET imaging of endogenous CD8+ T cells in vivo. Proc Natl Acad Sci U S A 2014; 111:1108-13. [PMID: 24390540 DOI: 10.1073/pnas.1316922111] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The noninvasive detection and quantification of CD8(+) T cells in vivo are important for both the detection and staging of CD8(+) lymphomas and for the monitoring of successful cancer immunotherapies, such as adoptive cell transfer and antibody-based immunotherapeutics. Here, antibody fragments are constructed to target murine CD8 to obtain rapid, high-contrast immuno-positron emission tomography (immuno-PET) images for the detection of CD8 expression in vivo. The variable regions of two anti-murine CD8-depleting antibodies (clones 2.43 and YTS169.4.2.1) were sequenced and reformatted into minibody (Mb) fragments (scFv-CH3). After production and purification, the Mbs retained their antigen specificity and bound primary CD8(+) T cells from the thymus, spleen, lymph nodes, and peripheral blood. Importantly, engineering of the parental antibodies into Mbs abolished the ability to deplete CD8(+) T cells in vivo. The Mbs were subsequently conjugated to S-2-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid for (64)Cu radiolabeling. The radiotracers were injected i.v. into antigen-positive, antigen-negative, immunodeficient, antigen-blocked, and antigen-depleted mice to evaluate specificity of uptake in lymphoid tissues by immuno-PET imaging and ex vivo biodistribution. Both (64)Cu-radiolabeled Mbs produced high-contrast immuno-PET images 4 h postinjection and showed specific uptake in the spleen and lymph nodes of antigen-positive mice.
Collapse
|
16
|
Bhatnagar P, Li Z, Choi Y, Guo J, Li F, Lee DY, Figliola M, Huls H, Lee DA, Zal T, Li KC, Cooper LJN. Imaging of genetically engineered T cells by PET using gold nanoparticles complexed to Copper-64. Integr Biol (Camb) 2013; 5:231-8. [PMID: 23034721 DOI: 10.1039/c2ib20093g] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adoptive transfer of primary T cells genetically modified to have desired specificity can exert an anti-tumor response in some patients. To improve our understanding of their therapeutic potential we have developed a clinically-appealing approach to reveal their in vivo biodistribution using nanoparticles that serve as a radiotracer for imaging by positron emission tomography (PET). T cells electroporated with DNA plasmids from the Sleeping Beauty transposon-transposase system to co-express a chimeric antigen receptor (CAR) specific for CD19 and Firefly luciferase (ffLuc) were propagated on CD19(+) K562-derived artificial antigen presenting cells. The approach to generating our clinical-grade CAR(+) T cells was adapted for electro-transfer of gold nanoparticles (GNPs) functionalized with (64)Cu(2+) using the macrocyclic chelator (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid, DOTA) and polyethyleneglycol (GNP-(64)Cu/PEG2000). MicroPET/CT was used to visualize CAR(+)EGFPffLucHyTK(+)GNP-(64)Cu/PEG2000(+) T cells and correlated with bioluminescence imaging. These data demonstrate that GNPs conjugated with (64)Cu(2+) can be prepared as a radiotracer for PET and used to image T cells using an approach that has translational implications.
Collapse
Affiliation(s)
- Parijat Bhatnagar
- Baylor College of Medicine & Texas Children's Hospital, Department of Obstetrics & Gynecology, Division of Maternal Fetal Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Buck D, Förschler A, Lapa C, Schuster T, Vollmar P, Korn T, Nessler S, Stadelmann C, Drzezga A, Buck AK, Wester HJ, Zimmer C, Krause BJ, Hemmer B. 18F-FDG PET Detects Inflammatory Infiltrates in Spinal Cord Experimental Autoimmune Encephalomyelitis Lesions. J Nucl Med 2012; 53:1269-76. [DOI: 10.2967/jnumed.111.102608] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
18
|
Johnström P, Fryer TD, Bird JL, Richards HK, Davenport AP. Dynamic in vivo imaging of receptors in small animals using positron emission tomography. Methods Mol Biol 2012; 897:221-37. [PMID: 22674168 DOI: 10.1007/978-1-61779-909-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Positron emission tomography (PET) is a functional imaging technique with the potential to image and quantify receptors in vivo with high sensitivity. PET has been used extensively to study major neurotransmitters such as dopamine, serotonin, and benzodiazepine in humans as well as proving to be a very powerful tool to accelerate development and assessment of existing and novel drugs. With the recent development of dedicated PET scanners for small animals, such as the microPET, it is now possible to perform functional imaging in small animals such as rodents at high resolution. This will allow the study of animal models of disease and longitudinal studies in these models to monitor disease progression or effect of treatment in the same animal. Furthermore, the complete pharmacokinetics of a drug as well as pharmacodynamic information can be obtained in a single animal. Thus, small animal imaging will significantly reduce the number of animals needed for this type of experiment as well as reducing the effect of inter-animal variation. Experimental protocols in small animal imaging potentially can be very labor intensive. In this chapter, we discuss methods and practical aspects related to this type of experiment using the microPET system.
Collapse
|
19
|
Homeostatic cytokines orchestrate the segregation of CD4 and CD8 memory T-cell reservoirs in mice. Blood 2011; 118:3039-50. [PMID: 21791416 DOI: 10.1182/blood-2011-04-349746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Memory T cells (T(M)s) have been detected in many tissues but their quantitative distribution remains largely undefined. We show that in mice there is a remarkably biased accumulation of long-term CD4 T(M)s into mucosal sites (mainly gut, especially Peyer patches), and CD8 T(M)s into lymph nodes and spleen (in particular, peripheral lymph nodes [PLNs]). This distinction correlates with their differentiated expression of PLN- and gut-homing markers. CD8 and CD4 T(M)s selectively require the expression of PLN-homing marker CCR7 or gut-homing marker α4β7 for maintenance. PLNs and gut supply CD8 and CD4 T(M)s with their individually favored homeostatic cytokine, IL-15, or IL-7. Cytokine stimulation in turn regulates the different gut-homing marker expression on CD4 and CD8 T(M)s. IL-15 plays a major role in vivo regulating CD8 T(M)s homing to PLNs. Thus, the reservoir segregation of CD4 and CD8 T(M)s meets their individual needs for homeostatic cytokines and is under feedback control of cytokine stimulation.
Collapse
|
20
|
Leukocyte-cancer cell fusion: initiator of the warburg effect in malignancy? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 714:151-72. [PMID: 21506013 DOI: 10.1007/978-94-007-0782-5_8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The causes of metastasis remain unknown, however it has been proposed for nearly a century that metastatic cells are generated by fusion of tumor cells with tumor-associated leukocytes such as macrophages. Indeed, regardless of cell or tissue origin, when cancer cells in the original in situ tumor transform to malignant, invasive cells, they generally become aneuploid and begin to express molecules and traits characteristic of activated macrophages. This includes two key features of malignancy: chemotactic motility and the use of aerobic glycolysis as a metabolic energy source (the Warburg effect). Here we review evidence that these phenomena can be well-explained by macrophage-cancer cell fusion, as evidenced by studies of experimental macrophage-melanoma hybrids generated in vitro and spontaneous host-tumor hybrids in animals and more recently humans. A key finding to emerge is that experimental and spontaneous cancer cell hybrids alike displayed a high degree of constitutive autophagy, a macrophage trait that is expressed under hypoxia and nutrient deprivation as part of the Warburg effect. Subsequent surveys of 21 different human cancers from nearly 2,000 cases recently revealed that the vast majority (~85%) exhibited autophagy and that this was associated with tumor proliferation and metastasis. While much work needs to be done, we posit that these findings with human cancers could be a reflection of widespread leukocyte-cancer cell fusion as an initiator of metastasis. Such fusions would generate hybrids that express the macrophage capabilities for motility and survival under adverse conditions of hypoxia and nutrient deprivation, while at the same time maintaining the deregulated mitotic cycle of the cancer cell fusion partner.
Collapse
|
21
|
Laing RE, Nair-Gill E, Witte ON, Radu CG. Visualizing cancer and immune cell function with metabolic positron emission tomography. Curr Opin Genet Dev 2010; 20:100-5. [PMID: 19931447 DOI: 10.1016/j.gde.2009.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 10/17/2009] [Indexed: 02/02/2023]
Abstract
Cancer cells and immune cells modulate their metabolism according to specific needs during cancer progression and immune responses. The ability to measure cellular metabolic function in vivo would enable the evaluation of tumors and their response to therapy and also the effectiveness of cellular immune responses to cancer. Positron emission tomography (PET) is a highly sensitive clinical imaging modality that enables whole-body, quantitative measurements of tissue biochemical function. Here, we review work using PET probes for specific metabolic pathways to measure cell function in cancer and immunity. We focus on the use of probes for glycolysis and nucleoside salvage and then discuss the development of new metabolic probes that visualize distinct parameters of cell function during disease.
Collapse
Affiliation(s)
- Rachel E Laing
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | | | | | | |
Collapse
|
22
|
Wang Q, Ornstein M, Kaufman HL. Imaging the immune response to monitor tumor immunotherapy. Expert Rev Vaccines 2009; 8:1427-37. [PMID: 19803763 DOI: 10.1586/erv.09.100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The goal of cancer immunotherapy is to promote antitumor immunity, and novel approaches include vaccination, adoptive transfer of tumor-reactive T cells, and administration of monoclonal antibodies and small molecules that target immune regulatory pathways. The molecular and cellular events responsible for tumor rejection are not completely defined and correlative studies have been used to help understand the mechanisms and extent of immune activation and tumor regression with these approaches. The real-time monitoring of immune responses to immunotherapy has been challenging as specific cell subsets may be difficult to define, and molecular pathways have evolved functionally diverse outcomes in different cells and in different tissues. Recently, improvements in optics and digital imaging have led to novel imaging techniques that make it possible to track the migration of individual immune cells ex vivo and in vivo, and to detect the dynamic interactions between T cells and antigen-presenting cells or tumor cells within complex microenvironments, including lymphoid tissue and established tumors. This review will explain some of the more established imaging techniques and discuss their role in monitoring the immune response in patients treated with various tumor immunotherapy approaches.
Collapse
Affiliation(s)
- Qin Wang
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA.
| | | | | |
Collapse
|
23
|
Inflammation on the mind: visualizing immunity in the central nervous system. Curr Top Microbiol Immunol 2009; 334:227-63. [PMID: 19521688 DOI: 10.1007/978-3-540-93864-4_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The central nervous system (CNS) is a remarkably complex structure that utilizes electrochemical signaling to coordinate activities throughout the entire body. Because the nervous system contains nonreplicative cells, it is postulated that, through evolutionary pressures, this compartment has acquired specialized mechanisms to limit damage. One potential source of damage comes from our immune system, which has the capacity to survey the CNS and periphery for the presence of foreign material. The immune system is equipped with numerous effector mechanisms and can greatly alter the homeostasis and function of the CNS. Degeneration, autoimmunity, and pathogen infection can all result in acute, and sometimes chronic, inflammation within the CNS. Understanding the specialized functionality of innate and adaptive immune cells within the CNS is critical to the design of more efficacious treatments to mitigate CNS inflammatory conditions. Much of our knowledge of CNS-immune interactions stems from seminal studies that have used static and dynamic imaging approaches to visualize inflammatory cells responding to different CNS conditions. This review will focus on how imaging techniques have elevated our understanding of CNS inflammation as well as the exciting prospects that lie ahead as we begin to pursue investigation of the inflamed CNS in real time.
Collapse
|
24
|
Abstract
Immune system activation can be elicited in viral infections, active immunization, or cancer immunotherapy, leading to the final common phenotype of increased glycolytic use by immune cells and subsequent detection by 18F-FDG PET. Because 18F-FDG is also used in baseline staging PET/CT scans and in tumor response assessment, physicians are faced with a unique challenge when evaluating tumor response in patients receiving cancer immunotherapy. The burgeoning field of cancer immunotherapy and the paucity of PET probes that can reliably differentiate activated immune cells from metabolically active cancer cells underscore the pressing need to identify and develop additional molecular imaging strategies. In an effort to address this concern, investigators have taken several molecular imaging approaches for cancer immunotherapy. Direct ex vivo labeling of T lymphocytes with radioactive probes before reinfusion represents the earliest attempts but has proven to be clinically limited because of significant PET probe dilution from proliferation of activated immune cells. Another approach is the indirect in vivo labeling of immune cells via PET reporter gene expression and involves the ex vivo genetic engineering of T lymphocytes with a reporter gene, reinfusion into the host, and the subsequent use of a PET probe specific for the reporter gene. The most recent approach involves the direct in vivo labeling of immune cells by targeting endogenous immune cell biochemical pathways that are differentially expressed during activation. In conclusion, these novel PET-based imaging approaches have demonstrated promise toward the goal of in vivo, noninvasive immune monitoring strategies for evaluating cancer immunotherapy.
Collapse
|