1
|
Gold L, Barci E, Brendel M, Orth M, Cheng J, Kirchleitner SV, Bartos LM, Pötter D, Kirchner MA, Unterrainer LM, Kaiser L, Ziegler S, Weidner L, Riemenschneider MJ, Unterrainer M, Belka C, Tonn JC, Bartenstein P, Niyazi M, von Baumgarten L, Kälin RE, Glass R, Lauber K, Albert NL, Holzgreve A. The Traumatic Inoculation Process Affects TSPO Radioligand Uptake in Experimental Orthotopic Glioblastoma. Biomedicines 2024; 12:188. [PMID: 38255293 PMCID: PMC10813339 DOI: 10.3390/biomedicines12010188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND The translocator protein (TSPO) has been proven to have great potential as a target for the positron emission tomography (PET) imaging of glioblastoma. However, there is an ongoing debate about the potential various sources of the TSPO PET signal. This work investigates the impact of the inoculation-driven immune response on the PET signal in experimental orthotopic glioblastoma. METHODS Serial [18F]GE-180 and O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) PET scans were performed at day 7/8 and day 14/15 after the inoculation of GL261 mouse glioblastoma cells (n = 24) or saline (sham, n = 6) into the right striatum of immunocompetent C57BL/6 mice. An additional n = 25 sham mice underwent [18F]GE-180 PET and/or autoradiography (ARG) at days 7, 14, 21, 28, 35, 50 and 90 in order to monitor potential reactive processes that were solely related to the inoculation procedure. In vivo imaging results were directly compared to tissue-based analyses including ARG and immunohistochemistry. RESULTS We found that the inoculation process represents an immunogenic event, which significantly contributes to TSPO radioligand uptake. [18F]GE-180 uptake in GL261-bearing mice surpassed [18F]FET uptake both in the extent and the intensity, e.g., mean target-to-background ratio (TBRmean) in PET at day 7/8: 1.22 for [18F]GE-180 vs. 1.04 for [18F]FET, p < 0.001. Sham mice showed increased [18F]GE-180 uptake at the inoculation channel, which, however, continuously decreased over time (e.g., TBRmean in PET: 1.20 at day 7 vs. 1.09 at day 35, p = 0.04). At the inoculation channel, the percentage of TSPO/IBA1 co-staining decreased, whereas TSPO/GFAP (glial fibrillary acidic protein) co-staining increased over time (p < 0.001). CONCLUSION We identify the inoculation-driven immune response to be a relevant contributor to the PET signal and add a new aspect to consider for planning PET imaging studies in orthotopic glioblastoma models.
Collapse
Affiliation(s)
- Lukas Gold
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Enio Barci
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- Munich Cluster for Systems Neurology (SyNergy), LMU Munich, 81377 Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Jiying Cheng
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Sabrina V. Kirchleitner
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr 15, 81377 Munich, Germany
| | - Laura M. Bartos
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Dennis Pötter
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Maximilian A. Kirchner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Lena Kaiser
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Sibylle Ziegler
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| | - Lorraine Weidner
- Department of Neuropathology, Regensburg University Hospital, 93053 Regensburg, Germany
| | | | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- DIE RADIOLOGIE, 80331 Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- Munich Cluster for Systems Neurology (SyNergy), LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Roland E. Kälin
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 81377 Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.G.)
| |
Collapse
|
2
|
Multi-Targeted Neutron Capture Therapy Combined with an 18 kDa Translocator Protein-Targeted Boron Compound Is an Effective Strategy in a Rat Brain Tumor Model. Cancers (Basel) 2023; 15:cancers15041034. [PMID: 36831378 PMCID: PMC9953932 DOI: 10.3390/cancers15041034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Boron neutron capture therapy (BNCT) has been adapted to high-grade gliomas (HG); however, some gliomas are refractory to BNCT using boronophenylalanine (BPA). In this study, the feasibility of BNCT targeting the 18 kDa translocator protein (TSPO) expressed in glioblastoma and surrounding environmental cells was investigated. METHODS Three rat glioma cell lines, an F98 rat glioma bearing brain tumor model, DPA-BSTPG which is a boron-10 compound targeting TSPO, BPA, and sodium borocaptate (BSH) were used. TSPO expression was evaluated in the F98 rat glioma model. Boron uptake was assessed in three rat glioma cell lines and in the F98 rat glioma model. In vitro and in vivo neutron irradiation experiments were performed. RESULTS DPA-BSTPG was efficiently taken up in vitro. The brain tumor has 16-fold higher TSPO expressions than its brain tissue. The compound biological effectiveness value of DPA-BSTPG was 8.43 to F98 rat glioma cells. The boron concentration in the tumor using DPA-BSTPG convection-enhanced delivery (CED) administration was approximately twice as high as using BPA intravenous administration. BNCT using DPA-BSTPG has significant efficacy over the untreated group. BNCT using a combination of BPA and DPA-BSTPG gained significantly longer survival times than using BPA alone. CONCLUSION DPA-BSTPG in combination with BPA may provide the multi-targeted neutron capture therapy against HG.
Collapse
|
3
|
Barca C, Foray C, Zinnhardt B, Winkeler A, Herrlinger U, Grauer OM, Jacobs AH. In Vivo Quantitative Imaging of Glioma Heterogeneity Employing Positron Emission Tomography. Cancers (Basel) 2022; 14:cancers14133139. [PMID: 35804911 PMCID: PMC9264799 DOI: 10.3390/cancers14133139] [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: 05/19/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma is the most common primary brain tumor, highly aggressive by being proliferative, neovascularized and invasive, heavily infiltrated by immunosuppressive glioma-associated myeloid cells (GAMs), including glioma-associated microglia/macrophages (GAMM) and myeloid-derived suppressor cells (MDSCs). Quantifying GAMs by molecular imaging could support patient selection for GAMs-targeting immunotherapy, drug target engagement and further assessment of clinical response. Magnetic resonance imaging (MRI) and amino acid positron emission tomography (PET) are clinically established imaging methods informing on tumor size, localization and secondary phenomena but remain quite limited in defining tumor heterogeneity, a key feature of glioma resistance mechanisms. The combination of different imaging modalities improved the in vivo characterization of the tumor mass by defining functionally distinct tissues probably linked to tumor regression, progression and infiltration. In-depth image validation on tracer specificity, biological function and quantification is critical for clinical decision making. The current review provides a comprehensive overview of the relevant experimental and clinical data concerning the spatiotemporal relationship between tumor cells and GAMs using PET imaging, with a special interest in the combination of amino acid and translocator protein (TSPO) PET imaging to define heterogeneity and as therapy readouts.
Collapse
Affiliation(s)
- Cristina Barca
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Correspondence: (C.B.); (A.H.J.)
| | - Claudia Foray
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Biomarkers & Translational Technologies (BTT), Pharma Research & Early Development (pRED), F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Alexandra Winkeler
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, F-91401 Orsay, France;
| | - Ulrich Herrlinger
- Division of Clinical Neuro-Oncology, Department of Neurology, University Hospital Bonn, D-53105 Bonn, Germany;
- Centre of Integrated Oncology (CIO), University Hospital Bonn, D-53127 Bonn, Germany
| | - Oliver M. Grauer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, D-48149 Münster, Germany;
| | - Andreas H. Jacobs
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Centre of Integrated Oncology (CIO), University Hospital Bonn, D-53127 Bonn, Germany
- Department of Geriatrics with Neurology, Johanniter Hospital, D-53113 Bonn, Germany
- Correspondence: (C.B.); (A.H.J.)
| |
Collapse
|
4
|
Zinnhardt B, Roncaroli F, Foray C, Agushi E, Osrah B, Hugon G, Jacobs AH, Winkeler A. Imaging of the glioma microenvironment by TSPO PET. Eur J Nucl Med Mol Imaging 2021; 49:174-185. [PMID: 33721063 DOI: 10.1007/s00259-021-05276-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Gliomas are highly dynamic and heterogeneous tumours of the central nervous system (CNS). They constitute the most common neoplasm of the CNS and the second most common cause of death from intracranial disease after stroke. The advances in detailing the genetic profile of paediatric and adult gliomas along with the progress in MRI and PET multimodal molecular imaging technologies have greatly improved prognostic stratification of patients with glioma and informed on treatment decisions. Amino acid PET has already gained broad clinical application in the study of gliomas. PET imaging targeting the translocator protein (TSPO) has recently been applied to decipher the heterogeneity and dynamics of the tumour microenvironment (TME) and its various cellular components especially in view of targeted immune therapies with the goal to delineate pro- and anti-glioma immune cell modulation. The current review provides a comprehensive overview on the historical developments of TSPO PET for gliomas and summarizes the most relevant experimental and clinical data with regard to the assessment and quantification of various cellular components with the TME of gliomas by in vivo TSPO PET imaging.
Collapse
Affiliation(s)
- Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster (WWU), Münster, Germany
- Biomarkers and Translational Technologies, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, Manchester, UK
| | - Claudia Foray
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster (WWU), Münster, Germany
| | - Erjon Agushi
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, Manchester, UK
| | - Bahiya Osrah
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, Manchester, UK
| | - Gaëlle Hugon
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Université Paris-Saclay, Orsay, France
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster (WWU), Münster, Germany
- Department of Geriatrics and Neurology, Johanniter Hospital, Bonn, Germany
| | - Alexandra Winkeler
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Université Paris-Saclay, Orsay, France.
- CEA, DRF, JOLIOT, SHFJ, Orsay, France.
| |
Collapse
|
5
|
Tran V, Lux F, Tournier N, Jego B, Maître X, Anisorac M, Comtat C, Jan S, Selmeczi K, Evans MJ, Tillement O, Kuhnast B, Truillet C. Quantitative Tissue Pharmacokinetics and EPR Effect of AGuIX Nanoparticles: A Multimodal Imaging Study in an Orthotopic Glioblastoma Rat Model and Healthy Macaque. Adv Healthc Mater 2021; 10:e2100656. [PMID: 34212539 DOI: 10.1002/adhm.202100656] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/29/2021] [Indexed: 01/10/2023]
Abstract
AGuIX are emerging radiosensitizing nanoparticles (NPs) for precision radiotherapy (RT) under clinical evaluation (Phase 2). Despite being accompanied by MRI thanks to the presence of gadolinium (Gd) at its surface, more sensitive and quantifiable imaging technique should further leverage the full potential of this technology. In this study, it is shown that 89 Zr can be labeled on such NPs directly for positron emission tomography (PET) imaging with a simple and scalable method. The stability of such complexes is remarkable in vitro and in vivo. Using a glioblastoma orthotopic rat model, it is shown that injected 89 Zr-AGuIX is detectable inside the tumor for at least 1 week. Interestingly, the particles seem to efficiently infiltrate the tumor even in necrotic areas, which places great hope for the treatment of radioresistant tumor. Lastly, the first PET/MR whole-body imaging is performed in non-human primate (NHP), which further demonstrates the translational potential of these bimodal NP.
Collapse
Affiliation(s)
- Vu‐Long Tran
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - François Lux
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
- Institut Universitaire de France (IUF) Paris France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Benoit Jego
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Xavier Maître
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Claude Comtat
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Sébastien Jan
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Michael J. Evans
- Department of Radiology and Biomedical Imaging University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Department of Pharmaceutical Chemistry University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Helen Diller Family Comprehensive Cancer Center University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
| | - Olivier Tillement
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
| | - Bertrand Kuhnast
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Charles Truillet
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| |
Collapse
|
6
|
Vettermann FJ, Harris S, Schmitt J, Unterrainer M, Lindner S, Rauchmann BS, Palleis C, Weidinger E, Beyer L, Eckenweber F, Schuster S, Biechele G, Ferschmann C, Milenkovic VM, Wetzel CH, Rupprecht R, Janowitz D, Buerger K, Perneczky R, Höglinger GU, Levin J, Haass C, Tonn JC, Niyazi M, Bartenstein P, Albert NL, Brendel M. Impact of TSPO Receptor Polymorphism on [ 18F]GE-180 Binding in Healthy Brain and Pseudo-Reference Regions of Neurooncological and Neurodegenerative Disorders. Life (Basel) 2021; 11:484. [PMID: 34073557 PMCID: PMC8229996 DOI: 10.3390/life11060484] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
TSPO-PET tracers are sensitive to a single-nucleotide polymorphism (rs6971-SNP), resulting in low-, medium- and high-affinity binders (LABs, MABs and HABS), but the clinical relevance of [18F]GE-180 is still unclear. We evaluated the impact of rs6971-SNP on in vivo [18F]GE-180 binding in a healthy brain and in pseudo-reference tissue in neuro-oncological and neurodegenerative diseases. Standardized uptake values (SUVs) of [18F]GE-180-PET were assessed using a manually drawn region of interest in the frontoparietal and cerebellar hemispheres. The SUVs were compared between the LABs, MABs and HABs in control, glioma, four-repeat tauopathy (4RT) and Alzheimer's disease (AD) subjects. Second, the SUVs were compared between the patients and controls within their rs6971-subgroups. After excluding patients with prior therapy, 24 LABs (7 control, 5 glioma, 6 4RT and 6 AD) were analyzed. Age- and sex-matched MABs (n = 38) and HABs (n = 50) were selected. The LABs had lower frontoparietal and cerebellar SUVs when compared with the MABs and HABs, but no significant difference was observed between the MABs and HABs. Within each rs6971 group, no SUV difference between the patients and controls was detected in the pseudo-reference tissues. The rs6971-SNP affects [18F]GE-180 quantification, revealing lower binding in the LABs when compared to the MABs and HABs. The frontoparietal and cerebellar ROIs were successfully validated as pseudo-reference regions.
Collapse
Affiliation(s)
- Franziska J Vettermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Julia Schmitt
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Marcus Unterrainer
- Department of Radiology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Radiology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Endy Weidinger
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Christian Ferschmann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College, London SW7 2AZ, UK
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg, Germany
| | - Joerg C Tonn
- Department of Neurosurgery, University Hospital of Munich, 81377 Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| |
Collapse
|
7
|
Zinnhardt B, Müther M, Roll W, Backhaus P, Jeibmann A, Foray C, Barca C, Döring C, Tavitian B, Dollé F, Weckesser M, Winkeler A, Hermann S, Wagner S, Wiendl H, Stummer W, Jacobs AH, Schäfers M, Grauer OM. TSPO imaging-guided characterization of the immunosuppressive myeloid tumor microenvironment in patients with malignant glioma. Neuro Oncol 2021; 22:1030-1043. [PMID: 32047908 DOI: 10.1093/neuonc/noaa023] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tumor-associated microglia and macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) are potent immunosuppressors in the glioma tumor microenvironment (TME). Their infiltration is associated with tumor grade, progression, and therapy resistance. Specific tools for image-guided analysis of spatiotemporal changes in the immunosuppressive myeloid tumor compartments are missing. We aimed (i) to evaluate the role of fluorodeoxyglucose (18F)DPA-714* (translocator protein [TSPO]) PET-MRI in the assessment of the immunosuppressive TME in glioma patients, and (ii) to cross-correlate imaging findings with in-depth immunophenotyping. METHODS To characterize the glioma TME, a mixed collective of 9 glioma patients underwent [18F]DPA-714-PET-MRI in addition to [18F]fluoro-ethyl-tyrosine (FET)-PET-MRI. Image-guided biopsy samples were immunophenotyped by multiparametric flow cytometry and immunohistochemistry. In vitro autoradiography was performed for image validation and assessment of tracer binding specificity. RESULTS We found a strong relationship (r = 0.84, P = 0.009) between the [18F]DPA-714 uptake and the number and activation level of glioma-associated myeloid cells (GAMs). TSPO expression was mainly restricted to human leukocyte antigen D related-positive (HLA-DR+) activated GAMs, particularly to tumor-infiltrating HLA-DR+ MDSCs and TAMs. [18F]DPA-714-positive tissue volumes exceeded [18F]FET-positive volumes and showed a differential spatial distribution. CONCLUSION [18F]DPA-714-PET may be used to non-invasively image the glioma-associated immunosuppressive TME in vivo. This imaging paradigm may also help to characterize the heterogeneity of the glioma TME with respect to the degree of myeloid cell infiltration at various disease stages. [18F]DPA-714 may also facilitate the development of new image-guided therapies targeting the myeloid-derived TME.
Collapse
Affiliation(s)
- Bastian Zinnhardt
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,Immune Image-IMI Consortium, University Hospital Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), University Hospital Münster, Münster, Germany
| | - Michael Müther
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Philipp Backhaus
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Astrid Jeibmann
- Institute of Neuroanatomy, University Hospital Münster, Münster, Germany
| | - Claudia Foray
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), University Hospital Münster, Münster, Germany
| | - Cristina Barca
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), University Hospital Münster, Münster, Germany
| | - Christian Döring
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Bertrand Tavitian
- Inserm Unit 970, Paris Cardiovascular Research Center, Paris, France
| | - Frédéric Dollé
- Inserm Unit 1023, In Vivo Molecular Imaging Laboratory, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Alexandra Winkeler
- Inserm Unit 1023, In Vivo Molecular Imaging Laboratory, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,Immune Image-IMI Consortium, University Hospital Münster, Münster, Germany
| | - Stefan Wagner
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Heinz Wiendl
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,Immune Image-IMI Consortium, University Hospital Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), University Hospital Münster, Münster, Germany.,Department of Geriatrics, Johanniter Hospital, Bonn, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging, University of Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,Immune Image-IMI Consortium, University Hospital Münster, Münster, Germany
| | - Oliver M Grauer
- Immune Image-IMI Consortium, University Hospital Münster, Münster, Germany.,Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| |
Collapse
|
8
|
Tripathi SK, Kean R, Bongiorno E, Hooper DC, Jin YY, Wickstrom E, McCue PA, Thakur ML. Targeting VPAC1 Receptors for Imaging Glioblastoma. Mol Imaging Biol 2021; 22:293-302. [PMID: 31292914 DOI: 10.1007/s11307-019-01388-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Scintigraphic imaging of malignant glioblastoma (MG) continues to be challenging. We hypothesized that VPAC1 cell surface receptors can be targeted for positron emission tomography (PET) imaging of orthotopically implanted MG in a mouse model, using a VPAC1-specific peptide [64Cu]TP3805. PROCEDURES The expression of VPAC1 in mouse GL261 and human U87 glioma cell lines was determined by western blot. The ability of [64Cu]TP3805 to bind to GL261 and U87 cells was studied by cell-binding. Receptor-blocking studies were performed to validate receptor specificity. GL261 tumors were implanted orthotopically in syngeneic T-bet knockout C57BL/6 mouse brain (N = 15) and allowed to grow for 2-3 weeks. Mice were injected i.v., first with ~ 150 μCi of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) then 24 h later with ~ 200 μCi of [64Cu]TP3805. In another set of tumor-bearing mice, (N = 5), ionic [64Cu]Cl2 was injected as a control. Mice were imaged at a 2-h post-injection using an Inveon micro-PET/CT, sacrificed and % ID/g of [64Cu]TP3805 and [64Cu]Cl2 were calculated in a tumor, normal brain, and other tissues. For histologic tissue examination, 3-μm thick sections of the tumors and normal brain were prepared, digital autoradiography (DAR) was performed, and then the sections were H&E stained for histologic examination. RESULTS Western blots showed a strong signal for VPAC1 on both cell lines. [64Cu]TP3805 cell-binding was 87 ± 1.5 %. Receptor-blocking reduced cell-binding to 24.3 ± 1.5 % (P < 0.01). PET imaging revealed remarkable accumulation of [64Cu]TP3805 in GL261 MG with a negligible background in the normal brain, as compared to [18F]FDG. Micro-PET/CT image analyses and tissue distribution showed that the brain tumor uptake for [64Cu]TP3805 was 8.2 ± 1.7 % ID/g and for [64Cu]Cl2 2.1 ± 0.5 % ID/g as compared to 1.0 ± 0.3 % ID/g and 1.4 ± 0.3 % ID/g for normal mouse brains, respectively. The high tumor/normal brain ratio for [64Cu]TP3805 (8.1 ± 1.1) allowed tumors to be visualized unequivocally. Histology and [64Cu]TP3805 DAR differentiated malignant tumors from healthy brain and confirmed PET findings. CONCLUSION Targeting VPAC1 receptors using [64Cu]TP3805 for PET imaging of MG is a promising novel approach and calls for further investigation.
Collapse
Affiliation(s)
- Sushil K Tripathi
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emily Bongiorno
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Douglas C Hooper
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Eric Wickstrom
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiology, Laboratories of Radiopharmaceutical Research and Molecular Imaging, 1020 Locust Street, 359-JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
9
|
Foray C, Valtorta S, Barca C, Winkeler A, Roll W, Müther M, Wagner S, Gardner ML, Hermann S, Schäfers M, Grauer OM, Moresco RM, Zinnhardt B, Jacobs AH. Imaging temozolomide-induced changes in the myeloid glioma microenvironment. Theranostics 2021; 11:2020-2033. [PMID: 33500706 PMCID: PMC7797694 DOI: 10.7150/thno.47269] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Rationale: The heterogeneous nature of gliomas makes the development and application of novel treatments challenging. In particular, infiltrating myeloid cells play a role in tumor progression and therapy resistance. Hence, a detailed understanding of the dynamic interplay of tumor cells and immune cells in vivo is necessary. To investigate the complex interaction between tumor progression and therapy-induced changes in the myeloid immune component of the tumor microenvironment, we used a combination of [18F]FET (amino acid metabolism) and [18F]DPA-714 (TSPO, GAMMs, tumor cells, astrocytes, endothelial cells) PET/MRI together with immune-phenotyping. The aim of the study was to monitor temozolomide (TMZ) treatment response and therapy-induced changes in the inflammatory tumor microenvironment (TME). Methods: Eighteen NMRInu/nu mice orthotopically implanted with Gli36dEGFR cells underwent MRI and PET/CT scans before and after treatment with TMZ or DMSO (vehicle). Tumor-to-background (striatum) uptake ratios were calculated and areas of unique tracer uptake (FET vs. DPA) were determined using an atlas-based volumetric approach. Results: TMZ therapy significantly modified the spatial distribution and uptake of both tracers. [18F]FET uptake was significantly reduced after therapy (-53 ± 84%) accompanied by a significant decrease of tumor volume (-17 ± 6%). In contrast, a significant increase (61 ± 33%) of [18F]DPA-714 uptake was detected by TSPO imaging in specific areas of the tumor. Immunohistochemistry (IHC) validated the reduction in tumor volumes and further revealed the presence of reactive TSPO-expressing glioma-associated microglia/macrophages (GAMMs) in the TME. Conclusion: We confirm the efficiency of [18F]FET-PET for monitoring TMZ-treatment response and demonstrate that in vivo TSPO-PET performed with [18F]DPA-714 can be used to identify specific reactive areas of myeloid cell infiltration in the TME.
Collapse
|
10
|
Ammer LM, Vollmann-Zwerenz A, Ruf V, Wetzel CH, Riemenschneider MJ, Albert NL, Beckhove P, Hau P. The Role of Translocator Protein TSPO in Hallmarks of Glioblastoma. Cancers (Basel) 2020; 12:cancers12102973. [PMID: 33066460 PMCID: PMC7602186 DOI: 10.3390/cancers12102973] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The translocator protein (TSPO) has been under extensive investigation as a specific marker in positron emission tomography (PET) to visualize brain lesions following injury or disease. In recent years, TSPO is increasingly appreciated as a potential novel therapeutic target in cancer. In Glioblastoma (GBM), the most malignant primary brain tumor, TSPO expression levels are strongly elevated and scientific evidence accumulates, hinting at a pivotal role of TSPO in tumorigenesis and glioma progression. The aim of this review is to summarize the current literature on TSPO with respect to its role both in diagnostics and especially with regard to the critical hallmarks of cancer postulated by Hanahan and Weinberg. Overall, our review contributes to a better understanding of the functional significance of TSPO in Glioblastoma and draws attention to TSPO as a potential modulator of treatment response and thus an important factor that may influence the clinical outcome of GBM. Abstract Glioblastoma (GBM) is the most fatal primary brain cancer in adults. Despite extensive treatment, tumors inevitably recur, leading to an average survival time shorter than 1.5 years. The 18 kDa translocator protein (TSPO) is abundantly expressed throughout the body including the central nervous system. The expression of TSPO increases in states of inflammation and brain injury due to microglia activation. Not least due to its location in the outer mitochondrial membrane, TSPO has been implicated with a broad spectrum of functions. These include the regulation of proliferation, apoptosis, migration, as well as mitochondrial functions such as mitochondrial respiration and oxidative stress regulation. TSPO is frequently overexpressed in GBM. Its expression level has been positively correlated to WHO grade, glioma cell proliferation, and poor prognosis of patients. Several lines of evidence indicate that TSPO plays a functional part in glioma hallmark features such as resistance to apoptosis, invasiveness, and proliferation. This review provides a critical overview of how TSPO could regulate several aspects of tumorigenesis in GBM, particularly in the context of the hallmarks of cancer proposed by Hanahan and Weinberg in 2011.
Collapse
Affiliation(s)
- Laura-Marie Ammer
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
| | - Arabel Vollmann-Zwerenz
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, Ludwig Maximilians University of Munich, 81377 Munich, Germany;
| | - Christian H. Wetzel
- Molecular Neurosciences, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany;
| | | | - Nathalie L. Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, 81377 Munich, Germany;
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI) and Department Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Peter Hau
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany; (L.-M.A.); (A.V.-Z.)
- Correspondence:
| |
Collapse
|
11
|
Pigeon H, Pérès EA, Truillet C, Jego B, Boumezbeur F, Caillé F, Zinnhardt B, Jacobs AH, Le Bihan D, Winkeler A. TSPO-PET and diffusion-weighted MRI for imaging a mouse model of infiltrative human glioma. Neuro Oncol 2020; 21:755-764. [PMID: 30721979 DOI: 10.1093/neuonc/noz029] [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] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most devastating brain tumor. Despite the use of multimodal treatments, most patients relapse, often due to the highly invasive nature of gliomas. However, the detection of glioma infiltration remains challenging. The aim of this study was to assess advanced PET and MRI techniques for visualizing biological activity and infiltration of the tumor. METHODS Using multimodality imaging, we investigated [18F]DPA-714, a radiotracer targeting the 18 kDa translocator protein (TSPO), [18F]FET PET, non-Gaussian diffusion MRI (apparent diffusion coefficient, kurtosis), and the S-index, a composite diffusion metric, to detect tumor infiltration in a human invasive glioma model. In vivo imaging findings were confirmed by autoradiography and immunofluorescence. RESULTS Increased tumor-to-contralateral [18F]DPA-714 uptake ratios (1.49 ± 0.11) were found starting 7 weeks after glioma cell implantation. TSPO-PET allowed visualization of glioma infiltration into the contralateral hemisphere 2 weeks earlier compared with the clinically relevant biomarker for biological glioma activity [18F]FET. Diffusion-weighted imaging (DWI), in particular kurtosis, was more sensitive than standard T2-weighted MRI to detect differences between the glioma-bearing and the contralateral hemisphere at 5 weeks. Immunofluorescence data reflect in vivo findings. Interestingly, labeling for tumoral and stromal TSPO indicates a predominant expression of TSPO by tumor cells. CONCLUSION These results suggest that advanced PET and MRI methods, such as [18F]DPA-714 and DWI, may be superior to standard imaging methods to visualize glioma growth and infiltration at an early stage.
Collapse
Affiliation(s)
- Hayet Pigeon
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Elodie A Pérès
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France.,NeuroSpin, CEA/Université Paris-Saclay, Gif sur Yvette, France.,Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, Caen, France
| | - Charles Truillet
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Benoit Jego
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | | | - Fabien Caillé
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Bastian Zinnhardt
- EIMI and Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany
| | - Andreas H Jacobs
- EIMI and Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany.,Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| | - Denis Le Bihan
- NeuroSpin, CEA/Université Paris-Saclay, Gif sur Yvette, France
| | - Alexandra Winkeler
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| |
Collapse
|
12
|
Banati RB, Wilcox P, Xu R, Yin G, Si E, Son ET, Shimizu M, Holsinger RMD, Parmar A, Zahra D, Arthur A, Middleton RJ, Liu GJ, Charil A, Graeber MB. Selective, high-contrast detection of syngeneic glioblastoma in vivo. Sci Rep 2020; 10:9968. [PMID: 32561881 PMCID: PMC7305160 DOI: 10.1038/s41598-020-67036-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 05/19/2020] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma is a highly malignant, largely therapy-resistant brain tumour. Deep infiltration of brain tissue by neoplastic cells represents the key problem of diffuse glioma. Much current research focuses on the molecular makeup of the visible tumour mass rather than the cellular interactions in the surrounding brain tissue infiltrated by the invasive glioma cells that cause the tumour’s ultimately lethal outcome. Diagnostic neuroimaging that enables the direct in vivo observation of the tumour infiltration zone and the local host tissue responses at a preclinical stage are important for the development of more effective glioma treatments. Here, we report an animal model that allows high-contrast imaging of wild-type glioma cells by positron emission tomography (PET) using [18 F]PBR111, a selective radioligand for the mitochondrial 18 kDa Translocator Protein (TSPO), in the Tspo−/− mouse strain (C57BL/6-Tspotm1GuMu(GuwiyangWurra)). The high selectivity of [18 F]PBR111 for the TSPO combined with the exclusive expression of TSPO in glioma cells infiltrating into null-background host tissue free of any TSPO expression, makes it possible, for the first time, to unequivocally and with uniquely high biological contrast identify peri-tumoral glioma cell invasion at preclinical stages in vivo. Comparison of the in vivo imaging signal from wild-type glioma cells in a null background with the signal in a wild-type host tissue, where the tumour induces the expected TSPO expression in the host’s glial cells, illustrates the substantial extent of the peritumoral host response to the growing tumour. The syngeneic tumour (TSPO+/+) in null background (TSPO−/−) model is thus well suited to study the interaction of the tumour front with the peri-tumoral tissue, and the experimental evaluation of new therapeutic approaches targeting the invasive behaviour of glioblastoma.
Collapse
Affiliation(s)
- Richard B Banati
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia. .,Medical Imaging, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| | - Paul Wilcox
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Ran Xu
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Grace Yin
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Emily Si
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Eric Taeyoung Son
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Mauricio Shimizu
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - R M Damian Holsinger
- Molecular Neuroscience, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Arvind Parmar
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - David Zahra
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Andrew Arthur
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia.,Medical Imaging, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Arnaud Charil
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Manuel B Graeber
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
13
|
Kaleağasıoğlu F, Ali DM, Berger MR. Multiple Facets of Autophagy and the Emerging Role of Alkylphosphocholines as Autophagy Modulators. Front Pharmacol 2020; 11:547. [PMID: 32410999 PMCID: PMC7201076 DOI: 10.3389/fphar.2020.00547] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a highly conserved multistep process and functions as passage for degrading and recycling protein aggregates and defective organelles in eukaryotic cells. Based on the nature of these materials, their size and degradation rate, four types of autophagy have been described, i.e. chaperone mediated autophagy, microautophagy, macroautophagy, and selective autophagy. One of the major regulators of this process is mTOR, which inhibits the downstream pathway of autophagy following the activation of its complex 1 (mTORC1). Alkylphosphocholine (APC) derivatives represent a novel class of antineoplastic agents that inhibit the serine-threonine kinase Akt (i.e. protein kinase B), which mediates cell survival and cause cell cycle arrest. They induce autophagy through inhibition of the Akt/mTOR cascade. They interfere with phospholipid turnover and thus modify signaling chains, which start from the cell membrane and modulate PI3K/Akt/mTOR, Ras-Raf-MAPK/ERK and SAPK/JNK pathways. APCs include miltefosine, perifosine, and erufosine, which represent the first-, second- and third generation of this class, respectively. In a high fraction of human cancers, constitutively active oncoprotein Akt1 suppresses autophagy in vitro and in vivo. mTOR is a down-stream target for Akt, the activation of which suppresses autophagy. However, treatment with APC derivatives will lead to dephosphorylation (hence deactivation) of mTOR and thus induces autophagy. Autophagy is a double-edged sword and may result in chemotherapeutic resistance as well as cancer cell death when apoptotic pathways are inactive. APCs display differential autophagy induction capabilities in different cancer cell types. Therefore, autophagy-dependent cellular responses need to be well understood in order to improve the chemotherapeutic outcome.
Collapse
Affiliation(s)
- Ferda Kaleağasıoğlu
- Department of Pharmacology, Faculty of Medicine, Near East University, Mersin, Turkey
| | - Doaa M. Ali
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pharmacology and Experimental Therapeutics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Martin R. Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
14
|
Foray C, Barca C, Backhaus P, Schelhaas S, Winkeler A, Viel T, Schäfers M, Grauer O, Jacobs AH, Zinnhardt B. Multimodal Molecular Imaging of the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:71-87. [PMID: 32030648 DOI: 10.1007/978-3-030-35727-6_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The tumour microenvironment (TME) surrounding tumour cells is a highly dynamic and heterogeneous composition of immune cells, fibroblasts, precursor cells, endothelial cells, signalling molecules and extracellular matrix (ECM) components. Due to the heterogeneity and the constant crosstalk between the TME and the tumour cells, the components of the TME are important prognostic parameters in cancer and determine the response to novel immunotherapies. To improve the characterization of the TME, novel non-invasive imaging paradigms targeting the complexity of the TME are urgently needed.The characterization of the TME by molecular imaging will (1) support early diagnosis and disease follow-up, (2) guide (stereotactic) biopsy sampling, (3) highlight the dynamic changes during disease pathogenesis in a non-invasive manner, (4) help monitor existing therapies, (5) support the development of novel TME-targeting therapies and (6) aid stratification of patients, according to the cellular composition of their tumours in correlation to their therapy response.This chapter will summarize the most recent developments and applications of molecular imaging paradigms beyond FDG for the characterization of the dynamic molecular and cellular changes in the TME.
Collapse
Affiliation(s)
- Claudia Foray
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), Münster, Germany
| | - Cristina Barca
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), Münster, Germany
| | - Philipp Backhaus
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany
| | - Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Alexandra Winkeler
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Thomas Viel
- Paris Centre de Recherche Cardiovasculaire, INSERM-U970, Université Paris Descartes, Paris, France
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany
| | - Oliver Grauer
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,PET Imaging in Drug Design and Development (PET3D), Münster, Germany.,Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany. .,PET Imaging in Drug Design and Development (PET3D), Münster, Germany. .,Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany.
| |
Collapse
|
15
|
Sinharay S, Tu TW, Kovacs ZI, Schreiber-Stainthorp W, Sundby M, Zhang X, Papadakis GZ, Reid WC, Frank JA, Hammoud DA. In vivo imaging of sterile microglial activation in rat brain after disrupting the blood-brain barrier with pulsed focused ultrasound: [18F]DPA-714 PET study. J Neuroinflammation 2019; 16:155. [PMID: 31345243 PMCID: PMC6657093 DOI: 10.1186/s12974-019-1543-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 12/26/2022] Open
Abstract
Background Magnetic resonance imaging (MRI)-guided pulsed focused ultrasound combined with the infusion of microbubbles (pFUS+MB) induces transient blood-brain barrier opening (BBBO) in targeted regions. pFUS+MB, through the facilitation of neurotherapeutics’ delivery, has been advocated as an adjuvant treatment for neurodegenerative diseases and malignancies. Sterile neuroinflammation has been recently described following pFUS+MB BBBO. In this study, we used PET imaging with [18F]-DPA714, a biomarker of translocator protein (TSPO), to assess for neuroinflammatory changes following single and multiple pFUS+MB sessions. Methods Three groups of Sprague-Dawley female rats received MRI-guided pFUS+MB (Optison™; 5–8 × 107 MB/rat) treatments to the left frontal cortex and right hippocampus. Group A rats were sonicated once. Group B rats were sonicated twice and group C rats were sonicated six times on weekly basis. Passive cavitation detection feedback (PCD) controlled the peak negative pressure during sonication. We performed T1-weighted scans immediately after sonication to assess efficiency of BBBO and T2*-weighted scans to evaluate for hypointense voxels. [18F]DPA-714 PET/CT scans were acquired after the BBB had closed, 24 h after sonication in group A and within an average of 10 days from the last sonication in groups B and C. Ratios of T1 enhancement, T2* values, and [18F]DPA-714 percent injected dose/cc (%ID/cc) values in the targeted areas to the contralateral brain were calculated. Histological assessment for microglial activation/astrocytosis was performed. Results In all groups, [18F]DPA-714 binding was increased at the sonicated compared to non-sonicated brain (%ID/cc ratios > 1). Immunohistopathology showed increased staining for microglial and astrocytic markers in the sonicated frontal cortex compared to contralateral brain and to a lesser extent in the sonicated hippocampus. Using MRI, we documented BBB disruption immediately after sonication with resolution of BBBO 24 h later. We found more T2* hypointense voxels with increasing number of sonications. In a longitudinal group of animals imaged after two and after six sonications, there was no cumulative increase of neuroinflammation on PET. Conclusion Using [18F]DPA-714 PET, we documented in vivo neuroinflammatory changes in association with pFUS+MB. Our protocol (utilizing PCD feedback to minimize damage) resulted in neuroinflammation visualized 24 h post one sonication. Our findings were supported by immunohistochemistry showing microglial activation and astrocytosis. Experimental sonication parameters intended for BBB disruption should be evaluated for neuroinflammatory sequelae prior to implementation in clinical trials. Electronic supplementary material The online version of this article (10.1186/s12974-019-1543-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanhita Sinharay
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.,University of Texas, MD Anderson Cancer Center, Houston, USA
| | - Tsang-Wei Tu
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology, Howard University, Washington DC, USA
| | - Zsofia I Kovacs
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Institute for Biomedical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - William Schreiber-Stainthorp
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Xiang Zhang
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Georgios Z Papadakis
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.,Department of Radiology, University of Crete and Department of Medical Imaging Heraklion University Hospital, Crete, Greece
| | - William C Reid
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Dima A Hammoud
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.
| |
Collapse
|
16
|
Comparison of 18F-GE-180 and dynamic 18F-FET PET in high grade glioma: a double-tracer pilot study. Eur J Nucl Med Mol Imaging 2018; 46:580-590. [DOI: 10.1007/s00259-018-4166-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022]
|
17
|
Dupont AC, Largeau B, Guilloteau D, Santiago Ribeiro MJ, Arlicot N. The Place of PET to Assess New Therapeutic Effectiveness in Neurodegenerative Diseases. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:7043578. [PMID: 29887768 PMCID: PMC5985069 DOI: 10.1155/2018/7043578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/01/2018] [Indexed: 12/16/2022]
Abstract
In vivo exploration of neurodegenerative diseases by positron emission tomography (PET) imaging has matured over the last 20 years, using dedicated radiopharmaceuticals targeting cellular metabolism, neurotransmission, neuroinflammation, or abnormal protein aggregates (beta-amyloid and intracellular microtubule inclusions containing hyperphosphorylated tau). The ability of PET to characterize biological processes at the cellular and molecular levels enables early detection and identification of molecular mechanisms associated with disease progression, by providing accurate, reliable, and longitudinally reproducible quantitative biomarkers. Thus, PET imaging has become a relevant imaging method for monitoring response to therapy, approved as an outcome measure in bioclinical trials. The aim of this paper is to review and discuss the current inputs of PET in the assessment of therapeutic effectiveness in neurodegenerative diseases connected by common pathophysiological mechanisms, including Parkinson's disease, Huntington's disease, dementia, amyotrophic lateral sclerosis, multiple sclerosis, and also in psychiatric disorders. We also discuss opportunities for PET imaging to drive more personalized neuroprotective and therapeutic strategies, taking into account individual variability, within the growing framework of precision medicine.
Collapse
Affiliation(s)
- Anne-Claire Dupont
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHRU de Tours, Unité de Radiopharmacie, Tours, France
| | | | - Denis Guilloteau
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHRU de Tours, Service de Médecine Nucléaire in vitro, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
| | - Maria Joao Santiago Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
- CHRU de Tours, Service de Médecine Nucléaire in vivo, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CHRU de Tours, Unité de Radiopharmacie, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
| |
Collapse
|
18
|
Induction of ER and mitochondrial stress by the alkylphosphocholine erufosine in oral squamous cell carcinoma cells. Cell Death Dis 2018; 9:296. [PMID: 29463797 PMCID: PMC5833417 DOI: 10.1038/s41419-018-0342-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/04/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Endoplasmic reticulum (ER) plays an essential role in cell function and survival. Accumulation of unfolded or misfolded proteins in the lumen of the ER activates the unfolded protein response (UPR), resulting in ER stress and subsequent apoptosis. The alkylphosphocholine erufosine is a known Akt-mTOR inhibitor in oral squamous cell carcinoma (OSCC). In the present study, we evaluate erufosine’s role to induce ER and mitochondrial stress leading to autophagy, apoptosis, and ROS induction. The cellular toxicity of erufosine was determined in two OSCC cell lines and gene expression and enrichment analyses were performed. A positive enrichment of ER stress upon erufosine exposure was observed, which was verified at protein levels for the ER stress sensors and their downstream mediators. Knockdown and pharmacological inhibition of the ER stress sensors PERK and XBP1 revealed their involvement into erufosine’s cellular effects, including proliferation, apoptosis, and autophagy induction. Autophagy was confirmed by increased acidic vacuoles and LC3-B levels. Upon erufosine exposure, calcium influx into the cytoplasm of the two OSCC cell lines was seen. Apoptosis was confirmed by nuclear staining, Annexin-V, and immunoblotting of caspases. The induction of mitochondrial stress upon erufosine exposure was predicted by gene set enrichment analysis (GSEA) and shown by erufosine’s effect on mitochondrial membrane potential, ATP, and ROS production in OSCC cells. These data show that ER and mitochondrial targeting by erufosine represents a new facet of its mechanism of action as well as a promising new framework in the treatment of head and neck cancers.
Collapse
|
19
|
TSPO PET for glioma imaging using the novel ligand 18F-GE-180: first results in patients with glioblastoma. Eur J Nucl Med Mol Imaging 2017; 44:2230-2238. [DOI: 10.1007/s00259-017-3799-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022]
|
20
|
Foss CA, Liu L, Mease RC, Wang H, Pasricha P, Pomper MG. Imaging Macrophage Accumulation in a Murine Model of Chronic Pancreatitis with 125I-Iodo-DPA-713 SPECT/CT. J Nucl Med 2017; 58:1685-1690. [PMID: 28522739 DOI: 10.2967/jnumed.117.189571] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022] Open
Abstract
Pancreatitis remains a diagnostic challenge in patients with mild to moderate disease, with current imaging modalities being inadequate. Given the prominent macrophage infiltration in chronic pancreatitis, we hypothesized that 125I-iodo-DPA-713, a small-molecule radiotracer that specifically targets macrophages, could be used with SPECT/CT to image pancreatic inflammation in a relevant experimental model. Methods: Chronic pancreatitis was induced with cerulein in C57BL/6 mice, which were contrasted with saline-injected control mice. The animals were imaged at 7 wk after induction using N,N-diethyl-2-(2-(3-125I-iodo-4-methoxyphenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide (125I-iodo-DPA-713) SPECT/CT or 18F-FDG PET/CT. The biodistribution of 125I-iodo-DPA-713 was determined under the same conditions, and a pair of mice was imaged using a fluorescent analog of 125I-iodo-DPA-713, DPA-713-IRDye800CW, for correlative histology. Results: Pancreatic 125I-iodo-DPA-713 uptake was significantly higher in treated mice than control mice (5.17% ± 1.18% vs. 2.41% ± 0.34% injected dose/g, P = 0.02), as corroborated by imaging. Mice imaged with 18F-FDG PET/CT showed cerulein-enhanced pancreatic uptake in addition to a moderate signal from healthy pancreas. Near-infrared fluorescence imaging with DPA-713-IRDye800CW showed strong pancreatic uptake, focal liver uptake, and gastrointestinal uptake in the treated mice, whereas the control mice showed only urinary excretion. Ex vivo fluorescence microscopy revealed a large influx of macrophages in the pancreas colocalizing with the retained fluorescent probe in the treated but not the control mice. Conclusion: These data support the application of both 125I-iodo-DPA-713 SPECT/CT and DPA-713-IRDye800CW near-infrared fluorescence to delineate pancreatic, liver, or intestinal inflammation in living mice.
Collapse
Affiliation(s)
- Catherine A Foss
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Liansheng Liu
- Center for Neurogastroenterology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ronnie C Mease
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Haofan Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Pankaj Pasricha
- Center for Neurogastroenterology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| |
Collapse
|
21
|
Zinnhardt B, Pigeon H, Thézé B, Viel T, Wachsmuth L, Fricke IB, Schelhaas S, Honold L, Schwegmann K, Wagner S, Faust A, Faber C, Kuhlmann MT, Hermann S, Schäfers M, Winkeler A, Jacobs AH. Combined PET Imaging of the Inflammatory Tumor Microenvironment Identifies Margins of Unique Radiotracer Uptake. Cancer Res 2017; 77:1831-1841. [PMID: 28137769 DOI: 10.1158/0008-5472.can-16-2628] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/12/2017] [Accepted: 01/12/2017] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment is highly heterogeneous. For gliomas, the tumor-associated inflammatory response is pivotal to support growth and invasion. Factors of glioma growth, inflammation, and invasion, such as the translocator protein (TSPO) and matrix metalloproteinases (MMP), may serve as specific imaging biomarkers of the glioma microenvironment. In this study, noninvasive imaging by PET with [18F]DPA-714 (TSPO) and [18F]BR-351 (MMP) was used for the assessment of localization and quantification of the expression of TSPO and MMP. Imaging was performed in addition to established clinical imaging biomarker of active tumor volume ([18F]FET) in conjunction with MRI. We hypothesized that each imaging biomarker revealed distinct areas of the heterogeneous glioma tissue in a mouse model of human glioma. Tracers were found to be increased 1.4- to 1.7-fold, with [18F]FET showing the biggest volume as depicted by a thresholding-based, volumes of interest analysis. Tumor areas, which could not be detected by a single tracer and/or MRI parameter alone, were measured. Specific compartments of [18F]DPA-714 (14%) and [18F]BR-351 (11%) volumes along the tumor rim could be identified. [18F]DPA-714 (TSPO) and [18F]BR-351 (MMP) matched with histology. Glioma-associated microglia/macrophages (GAM) were identified as TSPO and MMP sources. Multitracer and multimodal molecular imaging approaches may allow us to gain important insights into glioma-associated inflammation (GAM, MMP). Moreover, this noninvasive technique enables characterization of the glioma microenvironment with respect to the disease-driving cellular compartments at the various disease stages. Cancer Res; 77(8); 1831-41. ©2017 AACR.
Collapse
Affiliation(s)
- Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany.
| | - Hayet Pigeon
- Imagerie Moléculaire In Vivo, Inserm, CEA, Univ. Paris Sud, CNRS, Université Paris Saclay, CEA - Service Hospitalier Frédéric Joliot, Orsay, France
| | - Benoit Thézé
- Imagerie Moléculaire In Vivo, Inserm, CEA, Univ. Paris Sud, CNRS, Université Paris Saclay, CEA - Service Hospitalier Frédéric Joliot, Orsay, France
| | - Thomas Viel
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany.,PARCC INSERM-U970, Université Paris Descartes, Paris, France
| | - Lydia Wachsmuth
- Department of Clinical Radiology, University Hospital Münster, Münster, Germany
| | - Inga B Fricke
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany
| | - Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany
| | - Lisa Honold
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany
| | - Katrin Schwegmann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany
| | - Stefan Wagner
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Andreas Faust
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, University Hospital Münster, Münster, Germany.,DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany
| | - Michael T Kuhlmann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany.,DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany
| | - Alexandra Winkeler
- Imagerie Moléculaire In Vivo, Inserm, CEA, Univ. Paris Sud, CNRS, Université Paris Saclay, CEA - Service Hospitalier Frédéric Joliot, Orsay, France
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-University Münster, Münster, Germany.,DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany.,Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| |
Collapse
|
22
|
Galldiks N, Law I, Pope WB, Arbizu J, Langen KJ. The use of amino acid PET and conventional MRI for monitoring of brain tumor therapy. Neuroimage Clin 2016; 13:386-394. [PMID: 28116231 PMCID: PMC5226808 DOI: 10.1016/j.nicl.2016.12.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/09/2016] [Accepted: 12/16/2016] [Indexed: 12/03/2022]
Abstract
Routine diagnostics and treatment monitoring of brain tumors is usually based on contrast-enhanced MRI. However, the capacity of conventional MRI to differentiate tumor tissue from posttherapeutic effects following neurosurgical resection, chemoradiation, alkylating chemotherapy, radiosurgery, and/or immunotherapy may be limited. Metabolic imaging using PET can provide relevant additional information on tumor metabolism, which allows for more accurate diagnostics especially in clinically equivocal situations. This review article focuses predominantly on the amino acid PET tracers 11C-methyl-l-methionine (MET), O-(2-[18F]fluoroethyl)-l-tyrosine (FET) and 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine (FDOPA) and summarizes investigations regarding monitoring of brain tumor therapy.
Collapse
Affiliation(s)
- Norbert Galldiks
- Dept. of Neurology, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| | - Ian Law
- Dept.of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Whitney B. Pope
- Dept. of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Javier Arbizu
- Dept. of Nuclear Medicine, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- Dept. of Nuclear Medicine, University of Aachen, Aachen, Germany
| |
Collapse
|
23
|
Evaluation of PET Imaging Performance of the TSPO Radioligand [18F]DPA-714 in Mouse and Rat Models of Cancer and Inflammation. Mol Imaging Biol 2016; 18:127-34. [PMID: 26194010 PMCID: PMC4722075 DOI: 10.1007/s11307-015-0877-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose Many radioligands have been explored for imaging the 18-kDa translocator protein (TSPO), a diagnostic and therapeutic target for inflammation and cancer. Here, we investigated the TSPO radioligand [18F]DPA-714 for positron emission tomography (PET) imaging of cancer and inflammation. Procedures [18F]DPA-714 PET imaging was performed in 8 mouse and rat models of breast and brain cancer and 4 mouse and rat models of muscular and bowel inflammation. Results [18F]DPA-714 showed different uptake levels in healthy organs and malignant tissues of mice and rats. Although high and displaceable [18F]DPA-714 binding is observed ex vivo, TSPO-positive PET imaging of peripheral lesions of cancer and inflammation in mice did not show significant lesion-to-background signal ratios. Slower [18F]DPA-714 metabolism and muscle clearance in mice compared to rats may explain the elevated background signal in peripheral organs in this species. Conclusion Although TSPO is an evolutionary conserved protein, inter- and intra-species differences call for further exploration of the pharmacological parameters of TSPO radioligands. Electronic supplementary material The online version of this article (doi:10.1007/s11307-015-0877-x) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Derivatives of the pyrazolo[1,5-a]pyrimidine acetamide DPA-713 as translocator protein (TSPO) ligands and pro-apoptotic agents in human glioblastoma. Eur J Pharm Sci 2016; 96:186-192. [PMID: 27658888 DOI: 10.1016/j.ejps.2016.09.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 09/09/2016] [Accepted: 09/17/2016] [Indexed: 12/13/2022]
Abstract
The 18kDa translocator protein (TSPO) is a target for novel glioblastoma therapies due to its upregulation in this cancer and relatively low levels of expression in the healthy cortex. The pyrazolo[1,5-a]pyrimidine acetamides, exemplified by DPA-713 and DPA-714, are a class of high affinity TSPO ligands with selectivity over the central benzodiazepine receptor. In this study we have explored the potential anti-glioblastoma activity of a library of DPA-713 and DPA-714 analogues, and investigated the effect of amending the alkyl ether chain on TSPO affinity and functional potential. All ligands demonstrated nanomolar affinity for TSPO, but showed diverse functional activity, for example DPA-713 and DPA-714 did not affect the proliferation or viability of human T98G glioblastoma cells, while the hexyl ether and benzyl ether derivatives decreased proliferation of T98G cells without affecting proliferation in human fetal glial SVGp12 cells. These ligands also induced apoptosis and dissipated T98G mitochondrial membrane potential. This suggests that the nature of the alkyl ether chain of pyrazolo[1,5-a]pyrimidine acetamides has little influence on TSPO affinity but is important for functional activity of this class of TSPO ligands.
Collapse
|
25
|
Alkyl ether lipids, ion channels and lipid raft reorganization in cancer therapy. Pharmacol Ther 2016; 165:114-31. [DOI: 10.1016/j.pharmthera.2016.06.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 05/26/2016] [Indexed: 12/21/2022]
|
26
|
Roncaroli F, Su Z, Herholz K, Gerhard A, Turkheimer FE. TSPO expression in brain tumours: is TSPO a target for brain tumour imaging? Clin Transl Imaging 2016; 4:145-156. [PMID: 27077069 PMCID: PMC4820497 DOI: 10.1007/s40336-016-0168-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
Positron emission tomography (PET) alone or in combination with MRI is increasingly assuming a central role in the development of diagnostic and therapeutic strategies for brain tumours with the aim of addressing tumour heterogeneity, assisting in patient stratification, and contributing to predicting treatment response. The 18 kDa translocator protein (TSPO) is expressed in high-grade gliomas, while its expression is comparatively low in normal brain. In addition, the evidence of elevated TSPO in neoplastic cells has led to studies investigating TSPO as a transporter of anticancer drugs for brain delivery and a selective target for tumour tissue. The TSPO therefore represents an ideal candidate for molecular imaging studies. Knowledge of the biology of TSPO in normal brain cells, in-depth understanding of TSPO functions and biodistribution in neoplastic cells, accurate methods for quantification of uptake of TSPO tracers and pharmacokinetic data regarding TSPO-targeted drugs are required before introducing TSPO PET and TSPO-targeted treatment in clinical practice. In this review, we will discuss the impact of preclinical PET studies and the application of TSPO imaging in human brain tumours, the advantages and disadvantages of TSPO imaging compared to other imaging modalities and other PET tracers, and pathology studies on the extent and distribution of TSPO in gliomas. The suitability of TSPO as molecular target for treatment of brain tumours will also be the appraised.
Collapse
Affiliation(s)
- Federico Roncaroli
- Wolfson Molecular Imaging Centre, The University of Manchester, 7 Palatine Road, Withington, Manchester, M20 3LJ UK
| | - Zhangjie Su
- Wolfson Molecular Imaging Centre, The University of Manchester, 7 Palatine Road, Withington, Manchester, M20 3LJ UK
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, The University of Manchester, 7 Palatine Road, Withington, Manchester, M20 3LJ UK
| | - Alexander Gerhard
- Wolfson Molecular Imaging Centre, The University of Manchester, 7 Palatine Road, Withington, Manchester, M20 3LJ UK
| | | |
Collapse
|
27
|
Li W, Li X, Xu S, Ma X, Zhang Q. Expression of Tim4 in Glioma and its Regulatory Role in LN-18 Glioma Cells. Med Sci Monit 2016; 22:77-82. [PMID: 26741116 PMCID: PMC4710195 DOI: 10.12659/msm.894963] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Tim4 is a transmembrane protein known as T cell immunoglobulin and mucin domain containing protein-4. We speculated that Tim4 might be associated with glioma. This study aimed to investigate the expression level of Tim4 in gliomas and the regulatory role of Tim4 on the growth and apoptosis of LN-18 glioma cells. Material/Methods Tumor tissues and adjacent normal tissues were collected from patients with glioma. The expression level of Tim4 mRNA and protein was determined by RT-PCR and Western blot analyses, respectively to evaluate their association with glioma. Tim4 was overexpressed or silenced by siRNA interference in cultured human glioma cells LN-18. The growth and apoptosis of LN-18 cells was detected by MTT assay and flow cytometry. The colony-forming ability of LN-18 cells was assessed by the colony formation assay. The collection of human tissues was approved by the Research Ethics Committee at the Harbin Medical University Cancer Hospital and performed in strict accordance with international standards. All patients were required to sign the informed consent. Results The expression level of Tim4 mRNA and protein in tumor tissues was significantly higher compared with adjacent normal tissues. Antisense miRNA targeting Tim4 inhibited the growth of LN-18 cells, induced their apoptosis, and reduced their clonogenic capacity. In contrast, overexpression of Tim4 promoted the growth of LN-18 cells, inhibited their apoptosis, and enhanced their clonogenic potential. Conclusions The expression level of Tim-4 is closely associated with glioma. Decreased expression of Tim4 inhibited the growth and colony-forming ability of LN-18 cells and induced their apoptosis, whereas increased expression of Tim4 stimulated the growth and clonogenic potential of LN-18 cells and suppressed their apoptosis.
Collapse
Affiliation(s)
- Weiguo Li
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Shujun Xu
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Xiangyu Ma
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Qiujie Zhang
- Department of Health, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
28
|
Lee DE, Yue X, Ibrahim WG, Lentz MR, Peterson KL, Jagoda EM, Kassiou M, Maric D, Reid WC, Hammoud DA. Lack of neuroinflammation in the HIV-1 transgenic rat: an [(18)F]-DPA714 PET imaging study. J Neuroinflammation 2015; 12:171. [PMID: 26377670 PMCID: PMC4574011 DOI: 10.1186/s12974-015-0390-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/02/2015] [Indexed: 11/29/2022] Open
Abstract
Background HIV-associated neuroinflammation is believed to be a major contributing factor in the development of HIV-associated neurocognitive disorders (HAND). In this study, we used micropositron emission tomography (PET) imaging to quantify neuroinflammation in HIV-1 transgenic rat (Tg), a small animal model of HIV, known to develop neurological and behavioral problems. Methods Dynamic [18F]DPA-714 PET imaging was performed in Tg and age-matched wild-type (WT) rats in three age groups: 3-, 9-, and 16-month-old animals. As a positive control for neuroinflammation, we performed unilateral intrastriatal injection of quinolinic acid (QA) in a separate group of WT rats. To confirm our findings, we performed multiplex immunofluorescent staining for Iba1 and we measured cytokine/chemokine levels in brain lysates of Tg and WT rats at different ages. Results [18F]DPA-714 uptake in HIV-1 Tg rat brains was generally higher than in age-matched WT rats but this was not statistically significant in any age group. [18F]DPA-714 uptake in the QA-lesioned rats was significantly higher ipsilateral to the lesion compared to contralateral side indicating neuroinflammatory changes. Iba1 immunofluorescence showed no significant differences in microglial activation between the Tg and WT rats, while the QA-lesioned rats showed significant activation. Finally, cytokine/chemokine levels in brain lysates of the Tg rats and WT rats were not significantly different. Conclusion Microglial activation might not be the primary mechanism for neuropathology in the HIV-1 Tg rats. Although [18F]DPA-714 is a good biomarker of neuroinflammation, it cannot be reliably used as an in vivo biomarker of neurodegeneration in the HIV-1 Tg rat. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0390-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dianne E Lee
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wael G Ibrahim
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Margaret R Lentz
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Kristin L Peterson
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Elaine M Jagoda
- Molecular Imaging Program (MIP), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Michael Kassiou
- Chemistry Department, The University of Sydney, Sydney, Australia
| | - Dragan Maric
- Division of Intermural Research (DIR), National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - William C Reid
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA.
| |
Collapse
|
29
|
Zhang J. Mapping neuroinflammation in frontotemporal dementia with molecular PET imaging. J Neuroinflammation 2015; 12:108. [PMID: 26022249 PMCID: PMC4451729 DOI: 10.1186/s12974-015-0236-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/06/2015] [Indexed: 01/17/2023] Open
Abstract
Recent findings have led to a renewed interest and support for an active role of inflammation in neurodegenerative dementias and related neurologic disorders. Detection of neuroinflammation in vivo throughout the course of neurodegenerative diseases is of great clinical interest. Studies have shown that microglia activation (an indicator of neuroinflammation) may present at early stages of frontotemporal dementia (FTD), but the role of neuroinflammation in the pathogenesis of FTD is largely unknown. The first-generation translocator protein (TSPO) ligand ([11C]-PK11195) has been used to detect microglia activation in FTD, and the second-generation TSPO ligands have imaged neuroinflammation in vivo with improved pharmacokinetic properties. This paper reviews related literature and technical issues on mapping neuroinflammation in FTD with positron-emission tomography (PET) imaging. Early detection of neuroinflammation in FTD may identify new tools for diagnosis, novel treatment targets, and means to monitor therapeutic efficacy. More studies are needed to image and track neuroinflammation in FTD. It is anticipated that the advances of TSPO PET imaging will overcome technical difficulties, and molecular imaging of neuroinflammation will aid in the characterization of neuroinflammation in FTD. Such knowledge has the potential to shed light on the poorly understood pathogenesis of FTD and related dementias, and provide imaging markers to guide the development and assessment of new therapies.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, N6A 5A5, Canada.
| |
Collapse
|
30
|
Tsartsalis S, Dumas N, Tournier BB, Pham T, Moulin-Sallanon M, Grégoire MC, Charnay Y, Millet P. SPECT imaging of glioma with radioiodinated CLINDE: evidence from a mouse GL26 glioma model. EJNMMI Res 2015; 5:9. [PMID: 25853015 PMCID: PMC4385259 DOI: 10.1186/s13550-015-0092-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/24/2015] [Indexed: 12/30/2022] Open
Abstract
Background Recent research has demonstrated the potential of 18-kDa translocator protein (TSPO) to serve as a target for nuclear imaging of gliomas. The aim of this study was to evaluate SPECT imaging of GL26 mouse glioma using radioiodinated CLINDE, a TSPO-specific tracer. Methods GL26 cells, previously transfected with an enhanced green fluorescent protein (EGFP)-expressing lentivirus, were stereotactically implanted in the striatum of C57/Bl6 mice. At 4 weeks post-injection, dynamic SPECT scans with [123I]CLINDE were performed. A displacement study assessed specificity of tracer binding. SPECT images were compared to results of autoradiography, fluorescence microscopy, in situ nucleic acid hybridization, histology, and immunohistochemistry. Western blotting was performed to verify TSPO production by the tumor. Results Specific uptake of tracer by the tumor is observed with a high signal-to-noise ratio. Tracer uptake by the tumor is indeed 3.26 ± 0.32 times higher than that of the contralateral striatum, and 78% of the activity is displaceable by unlabeled CLINDE. Finally, TSPO is abundantly expressed by the GL26 cells. Conclusions The present study demonstrates the feasibility of [123I]CLINDE SPECT in translational studies and underlines its potential for clinical glioma SPECT imaging.
Collapse
Affiliation(s)
- Stergios Tsartsalis
- Vulnerability Biomarkers Unit, Division of General Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1225 Geneva, Chêne-Bourg Switzerland ; Department of Psychiatry, University of Geneva, 1 rue Michel-Servet, CH1211 Geneva 4, Switzerland
| | - Noé Dumas
- Vulnerability Biomarkers Unit, Division of General Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1225 Geneva, Chêne-Bourg Switzerland
| | - Benjamin B Tournier
- Vulnerability Biomarkers Unit, Division of General Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1225 Geneva, Chêne-Bourg Switzerland
| | - Tien Pham
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW 2234 Australia
| | | | - Marie-Claude Grégoire
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW 2234 Australia
| | - Yves Charnay
- Vulnerability Biomarkers Unit, Division of General Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1225 Geneva, Chêne-Bourg Switzerland
| | - Philippe Millet
- Vulnerability Biomarkers Unit, Division of General Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Chemin du Petit-Bel-Air 2, CH1225 Geneva, Chêne-Bourg Switzerland
| |
Collapse
|
31
|
Su YY, Yang GF, Lu GM, Wu S, Zhang LJ. PET and MR imaging of neuroinflammation in hepatic encephalopathy. Metab Brain Dis 2015; 30:31-45. [PMID: 25514861 DOI: 10.1007/s11011-014-9633-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/17/2014] [Indexed: 12/11/2022]
Abstract
Neurological or psychiatric abnormalities associated with hepatic encephalopathy (HE) range from subclinical findings to coma. HE is commonly accompanied with the accumulation of toxic substances in bloodstream. The toxicity effect of hyperammonemia on astrocyte, such as the alteration in neurotransmission, oxidative stress, astrocyte swelling, is considered as an important factor in the pathogenesis of HE. Besides, neuroinflammation has captured more attention in the process of HE, but the mechanism of neuroinflammation leading to HE remains unclear. Molecular imaging techniques such as positron emission tomography (PET) and magnetic resonance imaging (MRI) targeting activated microglia and/ or other mediators appear to be promising noninvasive approaches to assess HE. This review focuses on novel imaging and therapy strategies of neuroinflammation in HE.
Collapse
Affiliation(s)
- Yun Yan Su
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, 305 Zhongshan East Road, Xuanwu District, Nangjing, Jiangsu Province, 210002, China
| | | | | | | | | |
Collapse
|
32
|
Tang D, Nickels ML, Tantawy MN, Buck JR, Manning HC. Preclinical imaging evaluation of novel TSPO-PET ligand 2-(5,7-Diethyl-2-(4-(2-[(18)F]fluoroethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide ([ (18)F]VUIIS1008) in glioma. Mol Imaging Biol 2014; 16:813-20. [PMID: 24845529 PMCID: PMC4372299 DOI: 10.1007/s11307-014-0743-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Translocator protein (TSPO) concentrations are elevated in glioma, suggesting a role for TSPO positron emission tomography (PET) imaging in this setting. In preclinical PET studies, we evaluated a novel, high-affinity TSPO PET ligand, [(18)F]VUIIS1008, in healthy mice and glioma-bearing rats. PROCEDURES Dynamic PET data were acquired simultaneously with [(18)F]VUIIS1008 injection, with binding reversibility and specificity evaluated in vivo by non-radioactive ligand displacement or blocking. Compartmental analysis of PET data was performed using metabolite-corrected arterial input functions. Imaging was validated with histology and immunohistochemistry. RESULTS [(18)F]VUIIS1008 exhibited rapid uptake in TSPO-rich organs. PET ligand uptake was displaceable with non-radioactive VUIIS1008 or PBR06 in mice. Tumor accumulation of [(18)F]VUIIS1008 was blocked by pretreatment with VUIIS1008 in rats. [(18)F]VUIIS1008 exhibited improved tumor-to-background ratio and higher binding potential in tumors compared to a structurally similar pyrazolopyrimidine TSPO ligand, [(18)F]DPA-714. CONCLUSIONS The PET ligand [(18)F]VUIIS1008 exhibits promising characteristics as a tracer for imaging glioma. Further translational studies appear warranted.
Collapse
Affiliation(s)
- Dewei Tang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Michael L. Nickels
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - M. Noor Tantawy
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Jason R. Buck
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - H. Charles Manning
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| |
Collapse
|