1
|
Van Camp N, Lavisse S, Roost P, Gubinelli F, Hillmer A, Boutin H. TSPO imaging in animal models of brain diseases. Eur J Nucl Med Mol Imaging 2021; 49:77-109. [PMID: 34245328 PMCID: PMC8712305 DOI: 10.1007/s00259-021-05379-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022]
Abstract
Over the last 30 years, the 18-kDa TSPO protein has been considered as the PET imaging biomarker of reference to measure increased neuroinflammation. Generally assumed to image activated microglia, TSPO has also been detected in endothelial cells and activated astrocytes. Here, we provide an exhaustive overview of the recent literature on the TSPO-PET imaging (i) in the search and development of new TSPO tracers and (ii) in the understanding of acute and chronic neuroinflammation in animal models of neurological disorders. Generally, studies testing new TSPO radiotracers against the prototypic [11C]-R-PK11195 or more recent competitors use models of acute focal neuroinflammation (e.g. stroke or lipopolysaccharide injection). These studies have led to the development of over 60 new tracers during the last 15 years. These studies highlighted that interpretation of TSPO-PET is easier in acute models of focal lesions, whereas in chronic models with lower or diffuse microglial activation, such as models of Alzheimer's disease or Parkinson's disease, TSPO quantification for detection of neuroinflammation is more challenging, mirroring what is observed in clinic. Moreover, technical limitations of preclinical scanners provide a drawback when studying modest neuroinflammation in small brains (e.g. in mice). Overall, this review underlines the value of TSPO imaging to study the time course or response to treatment of neuroinflammation in acute or chronic models of diseases. As such, TSPO remains the gold standard biomarker reference for neuroinflammation, waiting for new radioligands for other, more specific targets for neuroinflammatory processes and/or immune cells to emerge.
Collapse
Affiliation(s)
- Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT, USA
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, M20 3LJ, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
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: 7.3] [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
|
3
|
Adhikari A, Singh P, Mahar KS, Adhikari M, Adhikari B, Zhang MR, Tiwari AK. Mapping of Translocator Protein (18 kDa) in Peripheral Sterile Inflammatory Disease and Cancer through PET Imaging. Mol Pharm 2021; 18:1507-1529. [PMID: 33645995 DOI: 10.1021/acs.molpharmaceut.1c00002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Positron emission tomography (PET) imaging of the translocator 18 kDa protein (TSPO) with radioligands has become an effective means of research in peripheral inflammatory conditions that occur in many diseases and cancers. The peripheral sterile inflammatory diseases (PSIDs) are associated with a diverse group of disorders that comprises numerous enduring insults including the cardiovascular, respiratory, gastrointestinal, or musculoskeletal system. TSPO has recently been introduced as a potential biomarker for peripheral sterile inflammatory diseases (PSIDs). The major critical issue related to PSIDs is its timely characterization and localization of inflammatory foci for proper therapy of patients. As an alternative to metabolic imaging, protein imaging expressed on immune cells after activation is of great importance. The five transmembrane domain translocator protein-18 kDa (TSPO) is upregulated on the mitochondrial cell surface of macrophages during inflammation, serving as a potential ligand for PET tracers. Additionally, the overexpressed TSPO protein has been positively correlated with various tumor malignancies. In view of the association of escalated TSPO expression in both disease conditions, it is an immensely important biomarker for PET imaging in oncology and PSIDs. In this review, we summarize the most outstanding advances on TSPO-targeted PSIDs and cancer in the development of TSPO ligands as a potential diagnostic tool, specifically discussing the last five years.
Collapse
Affiliation(s)
- Anupriya Adhikari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, (A Central University), Lucknow, Uttar Pradesh 226025, India
| | - Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, A Central University, Lucknow, Uttar Pradesh 226025, India
| | - Kamalesh S Mahar
- Birbal Sahni Institute of Palaeosciences, Lucknow, Uttar Pradesh 226007, India
| | - Manish Adhikari
- The George Washington University, Washington, D.C. 20052, United States
| | - Bhawana Adhikari
- Plasma Bio-science Research Center, Kwangwoon University, Seoul 01897, South Korea
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Anjani Kumar Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, (A Central University), Lucknow, Uttar Pradesh 226025, India
| |
Collapse
|
4
|
Tang D, Li J, Nickels ML, Huang G, Cohen AS, Manning HC. Preclinical Evaluation of a Novel TSPO PET Ligand 2-(7-Butyl-2-(4-(2-[ 18F]Fluoroethoxy)phenyl)-5-Methylpyrazolo[1,5-a]Pyrimidin-3-yl)-N,N-Diethylacetamide ( 18F-VUIIS1018A) to Image Glioma. Mol Imaging Biol 2019; 21:113-121. [PMID: 29869061 DOI: 10.1007/s11307-018-1198-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE There is an urgent need for the development of novel positron emission tomography (PET) tracers for glioma imaging. In this study, we developed a novel PET probe ([18F]VUIIS1018A) by targeting translocator protein (TSPO), an imaging biomarker for glioma. The purpose of this preclinical study was to evaluate this novel TSPO probe for glioma imaging. PROCEDURES In this study, we synthesized [19F]VUIIS1018A and the precursor for radiosynthesis of [18F]VUIIS1018A. TSPO binding affinity was confirmed using a radioligand competitive binding assay in C6 glioma cell lysate. Further, dynamic imaging studies were performed in rats using a microPET system. These studies include displacement and blocking studies for ligand reversibility and specificity evaluation, and compartment modeling of PET data for pharmacokinetic parameter measurement using metabolite-corrected arterial input functions and PMOD. RESULTS Compared to previously reported TSPO tracers including [18F]VUIIS1008 and [18F]DPA-714, the novel tracer [18F]VUIIS1018A demonstrated higher binding affinity and BPND. Pretreatment with the cold analog [19F]VUIIS1018A could partially block tumor accumulation of this novel tracer. Further, compartment modeling of this novel tracer also exhibited a greater tumor-to-background ratio, a higher tumor binding potential and a lower brain binding potential when compared with other TSPO probes, such as [18F]DPA-714 and [18F]VUIIS1008. CONCLUSIONS These studies illustrate that [18F]VUIIS1018A can serve as a promising TSPO PET tracer for glioma imaging and potentially imaging of other solid tumors.
Collapse
Affiliation(s)
- Dewei Tang
- Center for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Pudong New District, Shanghai, 200127, China
| | - Jun Li
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael L Nickels
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gang Huang
- Center for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Pudong New District, Shanghai, 200127, China
| | - Allison S Cohen
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA
| | - H Charles Manning
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA. .,Vanderbilt Center for Molecular Probes (CMP), Vanderbilt University Medical School, 1161 21st Ave. S., AA 1105 MCN, Nashville, TN, 37232-2310, USA. .,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA. .,Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, USA. .,Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA. .,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA. .,Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
5
|
Xia Y, Ledwitch K, Kuenze G, Duran A, Li J, Sanders CR, Manning C, Meiler J. A unified structural model of the mammalian translocator protein (TSPO). JOURNAL OF BIOMOLECULAR NMR 2019; 73:347-364. [PMID: 31243635 PMCID: PMC8006375 DOI: 10.1007/s10858-019-00257-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/10/2019] [Indexed: 05/10/2023]
Abstract
The translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor (PBR), is a membrane protein located on the outer mitochondrial membrane. Experimentally-derived structures of mouse TSPO (mTSPO) and its homologs from bacterial species have been determined by NMR spectroscopy and X-ray crystallography, respectively. These structures and ligand interactions within the TSPO binding pocket display distinct differences. Here, we leverage experimental and computational studies to derive a unified structural model of mTSPO in the presence and absence of the TSPO ligand, PK11195, and study the effects of DPC detergent micelles on the TSPO structure and ligand binding. From this work, we conclude that that the lipid-mimetic system used to solubilize mTSPO for NMR studies thermodynamically destabilizes the protein, introduces structural perturbations, and alters the characteristics of ligand binding. Furthermore, we used Rosetta to construct a unified mTSPO model that reconciles deviating features of the mammalian and bacterial TSPO. These deviating features are likely a consequence of the detergent system used for structure determination of mTSPO by NMR. The unified mTSPO model agrees with available experimental NMR data, appears to be physically realistic (i.e. thermodynamically not frustrated as judged by the Rosetta energy function), and simultaneously shares the structural features observed in sequence-conserved regions of the bacterial proteins. Finally, we identified the binding site for an imaging ligand VUIIS8310 that is currently positioned for clinical translation using NMR spectroscopy and propose a computational model of the VUIIS8310-mTSPO complex.
Collapse
Affiliation(s)
- Yan Xia
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Kaitlyn Ledwitch
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Georg Kuenze
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Amanda Duran
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jun Li
- Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Charles R Sanders
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37240, USA
| | - Charles Manning
- Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37240, USA.
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, MRBIII 5144B, Nashville, TN, 37232, USA.
| |
Collapse
|
6
|
Tang D, Li J, Buck JR, Tantawy MN, Xia Y, Harp JM, Nickels ML, Meiler J, Manning HC. Evaluation of TSPO PET Ligands [ 18F]VUIIS1009A and [ 18F]VUIIS1009B: Tracers for Cancer Imaging. Mol Imaging Biol 2018; 19:578-588. [PMID: 27853987 DOI: 10.1007/s11307-016-1027-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Positron emission tomography (PET) ligands targeting translocator protein (TSPO) are potential imaging diagnostics of cancer. In this study, we report two novel, high-affinity TSPO PET ligands that are 5,7 regioisomers, [18F]VUIIS1009A ([18F]3A) and [18F]VUIIS1009B ([18F]3B), and their initial in vitro and in vivo evaluation in healthy mice and glioma-bearing rats. PROCEDURES VUIIS1009A/B was synthesized and confirmed by X-ray crystallography. Interactions between TSPO binding pocket and novel ligands were evaluated and compared with contemporary TSPO ligands using 2D 1H-15N heteronuclear single quantum coherence (HSQC) spectroscopy. In vivo biodistribution of [18F]VUIIS1009A and [18F]VUIIS1009B was carried out in healthy mice with and without radioligand displacement. Dynamic PET imaging data were acquired simultaneously with [18F]VUIIS1009A/B injections in glioma-bearing rats, with binding reversibility and specificity evaluated by radioligand displacement. In vivo radiometabolite analysis was performed using radio-TLC, and quantitative analysis of PET data was performed using metabolite-corrected arterial input functions. Imaging was validated with histology and immunohistochemistry. RESULTS Both VUIIS1009A (3A) and VUIIS1009B (3B) were found to exhibit exceptional binding affinity to TSPO, with observed IC50 values against PK11195 approximately 500-fold lower than DPA-714. However, HSQC NMR suggested that VUIIS1009A and VUIIS1009B share a common binding pocket within mammalian TSPO (mTSPO) as DPA-714 and to a lesser extent, PK11195. [18F]VUIIS1009A ([18F]3A) and [18F]VUIIS1009B ([18F]3B) exhibited similar biodistribution in healthy mice. In rats bearing C6 gliomas, both [18F]VUIIS1009A and [18F]VUIIS1009B exhibited greater binding potential (k 3/k 4)in tumor tissue compared to [18F]DPA-714. Interestingly, [18F]VUIIS1009B exhibited significantly greater tumor uptake (V T) than [18F]VUIIS1009A, which was attributed primarily to greater plasma-to-tumor extraction efficiency. CONCLUSIONS The novel PET ligand [18F]VUIIS1009B exhibits promising characteristics for imaging glioma; its superiority over [18F]VUIIS1009A, a regioisomer, appears to be primarily due to improved plasma extraction efficiency. Continued evaluation of [18F]VUIIS1009B as a high-affinity TSPO PET ligand for precision medicine appears warranted.
Collapse
Affiliation(s)
- Dewei Tang
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jun Li
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Interdisciplinary Materials Science Program, Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37240, USA
| | - Jason R Buck
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Mohamed Noor Tantawy
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Yan Xia
- Center for Structural Biology (CSB), Vanderbilt University, Nashville, TN, 37205, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joel M Harp
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - Michael L Nickels
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jens Meiler
- Center for Structural Biology (CSB), Vanderbilt University, Nashville, TN, 37205, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute of Chemical Biology (VICB), Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - H Charles Manning
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA. .,Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, TN, 37232, USA. .,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA. .,Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
7
|
Buck JR, McKinley ET, Fu A, Abel TW, Thompson RC, Chambless L, Watchmaker JM, Harty JP, Cooper MK, Manning HC. Preclinical TSPO Ligand PET to Visualize Human Glioma Xenotransplants: A Preliminary Study. PLoS One 2015; 10:e0141659. [PMID: 26517124 PMCID: PMC4627825 DOI: 10.1371/journal.pone.0141659] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 10/12/2015] [Indexed: 11/18/2022] Open
Abstract
Current positron emission tomography (PET) imaging biomarkers for detection of infiltrating gliomas are limited. Translocator protein (TSPO) is a novel and promising biomarker for glioma PET imaging. To validate TSPO as a potential target for molecular imaging of glioma, TSPO expression was assayed in a tumor microarray containing 37 high-grade (III, IV) gliomas. TSPO staining was detected in all tumor specimens. Subsequently, PET imaging was performed with an aryloxyanilide-based TSPO ligand, [18F]PBR06, in primary orthotopic xenograft models of WHO grade III and IV gliomas. Selective uptake of [18F]PBR06 in engrafted tumor was measured. Furthermore, PET imaging with [18F]PBR06 demonstrated infiltrative glioma growth that was undetectable by traditional magnetic resonance imaging (MRI). Preliminary PET with [18F]PBR06 demonstrated a preferential tumor-to-normal background ratio in comparison to 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). These results suggest that TSPO PET imaging with such high-affinity radiotracers may represent a novel strategy to characterize distinct molecular features of glioma growth, as well as better define the extent of glioma infiltration for therapeutic purposes.
Collapse
Affiliation(s)
- Jason R. Buck
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Eliot T. McKinley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Allie Fu
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Ty W. Abel
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Reid C. Thompson
- Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Lola Chambless
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jennifer M. Watchmaker
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, United States of America
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - James P. Harty
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Michael K. Cooper
- Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, TN, United States of America
- Neurology Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States of America
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - H. Charles Manning
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, TN, United States of America
- Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
8
|
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: 2.7] [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
|
9
|
Austin CJD, Kahlert J, Kassiou M, Rendina LM. The translocator protein (TSPO): a novel target for cancer chemotherapy. Int J Biochem Cell Biol 2013; 45:1212-6. [PMID: 23518318 DOI: 10.1016/j.biocel.2013.03.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/21/2013] [Accepted: 03/05/2013] [Indexed: 01/02/2023]
Abstract
The translocator protein (TSPO) is an 18 kDa transmembrane protein primarily found in the outer mitochondrial membrane where it forms a key part of the mitochondrial permeability transition pore (MPTP). Omnipresent in almost all tissues, TSPO up-regulation has been connected to neuronal damage and inflammation, making the protein an important bio-imaging marker for disease progression. More recently, TSPO has attracted attention as a possible molecular target for tumour imaging and chemotherapy. In this review we summarize TSPO's molecular characteristics and highlight research progress in recent years in the field of TSPO-targeted cancer diagnostics and treatments.
Collapse
|
10
|
Tang D, Hight MR, McKinley ET, Fu A, Buck JR, Smith RA, Tantawy MN, Peterson TE, Colvin DC, Ansari MS, Nickels M, Manning HC. Quantitative preclinical imaging of TSPO expression in glioma using N,N-diethyl-2-(2-(4-(2-18F-fluoroethoxy)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide. J Nucl Med 2012; 53:287-94. [PMID: 22251555 DOI: 10.2967/jnumed.111.095653] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
UNLABELLED There is a critical need to develop and rigorously validate molecular imaging biomarkers to aid diagnosis and characterization of primary brain tumors. Elevated expression of translocator protein (TSPO) has been shown to predict disease progression and aggressive, invasive behavior in a variety of solid tumors. Thus, noninvasive molecular imaging of TSPO expression could form the basis of a novel, predictive cancer imaging biomarker. In quantitative preclinical PET studies, we evaluated a high-affinity pyrazolopyrimidinyl-based TSPO imaging ligand, N,N-diethyl-2-(2-(4-(2-(18)F-fluoroethoxy)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide ((18)F-DPA-714), as a translational probe for quantification of TSPO levels in glioma. METHODS Glioma-bearing rats were imaged with (18)F-DPA-714 in a small-animal PET system. Dynamic images were acquired simultaneously on injection of (18)F-DPA-714 (130-200 MBq/0.2 mL). Blood was collected to derive the arterial input function (AIF), with high-performance liquid chromatography radiometabolite analysis performed on selected samples for AIF correction. Compartmental modeling was performed using the corrected AIF. Specific tumor cell binding of DPA-714 was evaluated by radioligand displacement of (3)H-PK 11195 with DPA-714 in vitro and displacement of (18)F-DPA-714 with an excess of DPA-714 in vivo. Immediately after imaging, tumor and healthy brain tissues were harvested for validation by Western blotting and immunohistochemistry. RESULTS (18)F-DPA-714 was found to preferentially accumulate in tumors, with modest uptake in the contralateral brain. Infusion with DPA-714 (10 mg/kg) displaced (18)F-DPA-714 binding by greater than 60% on average. Tumor uptake of (18)F-DPA-714 was similar to another high-affinity TSPO imaging ligand, (18)F-N-fluoroacetyl-N-(2,5-dimethoxybenzyl)-2-phenoxyaniline, and agreed with ex vivo assay of TSPO levels in tumor and healthy brain. CONCLUSION These studies illustrate the feasibility of using (18)F-DPA-714 for visualization of TSPO-expressing brain tumors. Importantly, (18)F-DPA-714 appears suitable for quantitative assay of tumor TSPO levels in vivo. Given the relationship between elevated TSPO levels and poor outcome in oncology, these studies suggest the potential of (18)F-DPA-714 PET to serve as a novel predictive cancer imaging modality.
Collapse
Affiliation(s)
- Dewei Tang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Buck JR, McKinley ET, Hight MR, Fu A, Tang D, Smith RA, Tantawy MN, Peterson TE, Colvin D, Ansari MS, Baldwin RM, Zhao P, Guleryuz S, Manning HC. Quantitative, preclinical PET of translocator protein expression in glioma using 18F-N-fluoroacetyl-N-(2,5-dimethoxybenzyl)-2-phenoxyaniline. J Nucl Med 2010; 52:107-14. [PMID: 21149488 DOI: 10.2967/jnumed.110.081703] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Translocator protein (TSPO), also referred to as peripheral benzodiazepine receptor (PBR), is a crucial 18-kDa outer mitochondrial membrane protein involved in numerous cellular functions, including the regulation of cholesterol metabolism, steroidogenesis, and apoptosis. Elevated expression of TSPO in oncology correlates with disease progression and poor survival, suggesting that molecular probes capable of assaying TSPO levels may have potential as cancer imaging biomarkers. In preclinical PET studies, we characterized a high-affinity aryloxyanilide-based TSPO imaging ligand, 18F-N-fluoroacetyl-N-(2,5-dimethoxybenzyl)-2-phenoxyaniline (18F-PBR06), as a candidate probe for the quantitative assessment of TSPO expression in glioma. METHODS Glioma-bearing rats were imaged with 18F-PBR06 in a small-animal PET system. Dynamic images were acquired simultaneously on injection of 18F-PBR06 (70-100 MBq/0.2 mL). Over the course of scanning, arterial blood was collected to derive the input function, with high-performance liquid chromatography radiometabolite analysis performed on selected samples for arterial input function correction. Compartmental modeling of the PET data was performed using the corrected arterial input function. Specific tumor cell binding of PBR06 was evaluated by radioligand displacement of 3H-PK 11195 with PBR06 in vitro and by displacement of 18F-PBR06 with excess PBR06 in vivo. Immediately after imaging, tumor tissue and adjacent healthy brain were harvested for assay of TSPO protein levels by Western blotting and immunohistochemistry. RESULTS 18F-PBR06 was found to preferentially accumulate in tumors, with modest uptake in the contralateral brain, facilitating excellent contrast between tumor and adjacent tissue. Infusion with PBR06 (10 mg/kg) displaced 18F-PBR06 binding by approximately 75%. The accumulation of 18F-PBR06 in tumor tissues and adjacent brain agreed with the ex vivo assay of TSPO protein levels by Western blotting and quantitative immunohistochemistry. CONCLUSION These preclinical studies illustrate that 18F-PBR06 is a promising tracer for visualization of TSPO-expressing tumors. Importantly, the close correlation between 18F-PBR06 uptake and TSPO expression in tumors and normal tissues, coupled with the high degree of displaceable binding from both tumors and the normal brain, represents a significant improvement over other TSPO imaging ligands previously evaluated in glioma. These data suggest the potential of 18F-PBR06 to elucidate the role of TSPO in oncology, as well as its potential development as a cancer imaging biomarker.
Collapse
Affiliation(s)
- Jason R Buck
- Vanderbilt University Institute of Imaging Science, Department of Radiologyh and Radiological Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2310, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Cappelli A, Valenti S, Mancini A, Giuliani G, Anzini M, Altieri S, Bortolussi S, Ferrari C, Clerici AM, Zonta C, Carraro F, Filippi I, Giorgi G, Donati A, Ristori S, Vomero S, Concas A, Biggio G. Carborane-Conjugated 2-Quinolinecarboxamide Ligands of the Translocator Protein for Boron Neutron Capture Therapy. Bioconjug Chem 2010; 21:2213-21. [DOI: 10.1021/bc100195s] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Andrea Cappelli
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Salvatore Valenti
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Alessandra Mancini
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Germano Giuliani
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Maurizio Anzini
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Saverio Altieri
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Silva Bortolussi
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Cinzia Ferrari
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Anna Maria Clerici
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Cecilia Zonta
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Fabio Carraro
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Irene Filippi
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Gianluca Giorgi
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Alessandro Donati
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Sandra Ristori
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Salvatore Vomero
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Alessandra Concas
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| | - Giovanni Biggio
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy, Dipartimento di Fisica Nucleare e Teorica, Università di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Pavia, Via Ugo Bassi 6, 27100 Pavia, Italy, Dipartimento di Scienze Chirurgiche, Laboratorio di Chirurgia Sperimentale, Università di Pavia, Via Ferrata 9, 27100 Pavia, Italy,
| |
Collapse
|
13
|
Denora N, Laquintana V, Trapani A, Lopedota A, Latrofa A, Gallo JM, Trapani G. Translocator protein (TSPO) ligand-Ara-C (cytarabine) conjugates as a strategy to deliver antineoplastic drugs and to enhance drug clinical potential. Mol Pharm 2010; 7:2255-69. [PMID: 20958082 DOI: 10.1021/mp100235w] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aim of this work was to evaluate TSPO ligand-Ara-C conjugation as an approach for the selective delivery of the antineoplastic agent to brain tumors as well as for overcome P-gp resistance induction observed for the majority of cytotoxic agents, enhancing the drug clinical potential. To this end, the novel N-imidazopyridinacetyl-Ara-C conjugates 3a-c, 10 and 15 have been prepared and evaluated for their cytotoxicity against glioma cell lines. In contrast to that observed for 3a-c and 10, the conjugate 15 resulted stable in both phosphate buffer and physiological medium. In all cases, the release of free Ara-C from hydrolyzed conjugates was checked by HPLC and ESI-MS analysis. Conjugates 10 and 15 displayed very high in vitro TSPO affinity and selectivity, and, hence, they may possess potential for targeted brain delivery. Due to the favorable features displayed by the conjugate 15, it was further evaluated on glioma cell lines, expressing high levels of TSPO, in the presence and in the absence of specific nucleoside transport (NT) inhibitors. In contrast to that observed for the free Ara-C, the presence of NT inhibitors did not reduce the cytotoxic activity of 15. Moreover, conjugate 15, as N(4)-acyl derivative of Ara-C, should be resistant to inactivation by cytidine deaminase, and it may possess enhanced propensity to target brain tumor cells characterized by a reduced expression of NTs. In addition, this conjugate behaves as a clear P-gp modulator and thereby may be useful to reverse MDR. Transport studies across the MDCKII-MDR1 monolayer indicated that conjugate 15 should overcome the BBB by transcellular pathway. All these features may be useful for enhancing the clinical potential of the nucleoside drug Ara-C.
Collapse
Affiliation(s)
- Nunzio Denora
- Dipartimento Farmaco-Chimico, Facoltà di Farmacia, Università degli Studi di Bari, Via Orabona 4, 70125 Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
14
|
Bertomeu T, Zvereff V, Ibrahim A, Zehntner SP, Aliaga A, Rosa-Neto P, Bedell BJ, Falardeau P, Gourdeau H. TLN-4601 peripheral benzodiazepine receptor (PBR/TSPO) binding properties do not mediate apoptosis but confer tumor-specific accumulation. Biochem Pharmacol 2010; 80:1572-9. [PMID: 20655882 DOI: 10.1016/j.bcp.2010.07.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/05/2010] [Accepted: 07/12/2010] [Indexed: 12/22/2022]
Abstract
TLN-4601 is a farnesylated dibenzodiazepinone isolated from Micromonospora sp. with an antiproliferative effect on several human cancer cell lines. Although the mechanism of action of TLN-4601 is unknown, our earlier work indicated that TLN-4601 binds the PBR (peripheral benzodiazepine receptor; more recently known as the translocator protein or TSPO), an 18 kDa protein associated with the mitochondrial permeability transition (mPT) pore. While the exact function of the PBR remains a matter of debate, it has been implicated in heme and steroid synthesis, cellular growth and differentiation, oxygen consumption and apoptosis. Using the Jurkat immortalized T-lymphocyte cell line, documented to have negligible PBR expression, and Jurkat cells stably transfected with a human PBR cDNA, the present study demonstrates that TLN-4601 induces apoptosis independently of PBR expression. As PBRs are overexpressed in brain tumors compared to normal brain, we examined if TLN-4601 would preferentially accumulate in tumors using an intra-cerebral tumor model. Our results demonstrate the ability of TLN-4601 to effectively bind the PBR in vivo as determined by competitive binding assay and receptor occupancy. Analysis of TLN-4601 tissue and plasma indicated that TLN-4601 preferentially accumulates in the tumor. Indeed, drug levels were 200-fold higher in the tumor compared to the normal brain. TLN-4601 accumulation in the tumor (176 μg/g) was also significant compared to liver (24.8 μg/g; 7-fold) and plasma (16.2 μg/mL; 11-fold). Taken together our data indicate that while PBR binding does not mediate cell growth inhibition and apoptosis, PBR binding may allow for the specific accumulation of TLN-4601 in PBR positive tumors.
Collapse
Affiliation(s)
- T Bertomeu
- Thallion Pharmaceuticals Inc., 7150 Alexander-Fleming, Montréal, QC, H4S 2C8, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Binding of NIR-conPK and NIR-6T to astrocytomas and microglial cells: evidence for a protein related to TSPO. PLoS One 2009; 4:e8271. [PMID: 20020060 PMCID: PMC2792720 DOI: 10.1371/journal.pone.0008271] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 11/12/2009] [Indexed: 12/02/2022] Open
Abstract
PK 11195 and DAA1106 bind with high-affinity to the translocator protein (TSPO, formerly known as the peripheral benzodiazepine receptor). TSPO expression in glial cells increases in response to cytokines and pathological stimuli. Accordingly, [11C]-PK 11195 and [11C]-DAA1106 are recognized molecular imaging (MI) agents capable of monitoring changes in TSPO expression occurring in vivo and in response to various neuropathologies. Here we tested the pharmacological characteristics and TSPO-monitoring potential of two novel MI agents: NIR-conPK and NIR-6T. NIR-conPK is an analogue of PK 11195 conjugated to the near-infrared (NIR) emitting fluorophore: IRDye 800CW. NIR-6T is a DAA1106 analogue also conjugated to IRDye 800CW. We found that NIR-6T competed for [3H]-PK 11195 binding in astrocytoma cell homogenates with nanomolar affinity, but did not exhibit specific binding in intact astrocytoma cells in culture, indicating that NIR-6T is unlikely to constitute a useful MI agent for monitoring TSPO expression in intact cells. Conversely, we found that NIR-conPK did not compete for [3H]-PK 11195 binding in astrocytoma cell homogenate, but exhibited specific binding in intact astrocytoma cells in culture with nanomolar affinity, suggesting that NIR-conPK binds to a protein distinct, but related to, TSPO. Accordingly, treating intact astrocytoma cells and microglia in culture with cytokines led to significant changes in the amount of NIR-conPK specific binding without corresponding change in TSPO expression. Remarkably, the cytokine-induced changes in the protein targeted by NIR-conPK in intact microglia were selective, since IFN-γ (but not TNFα and TGFβ) increased the amount of NIR-conPK specific binding in these cells. Together these results suggest that NIR-conPK binds to a protein that is related to TSPO, and expressed by astrocytomas and microglia. Our results also suggest that the expression of this protein is increased by specific cytokines, and thus allows for the monitoring of a particular subtype of microglia activation.
Collapse
|
16
|
Molecular imaging of the translocator protein (TSPO) in a pre-clinical model of breast cancer. Mol Imaging Biol 2009; 12:349-58. [PMID: 19949989 DOI: 10.1007/s11307-009-0270-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 05/20/2009] [Accepted: 05/29/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE To quantitatively evaluate the utility of a translocator protein (TSPO)-targeted near-infrared (NIR) probe (NIR-conPK11195) for in vivo molecular imaging of TSPO in breast cancer. PROCEDURES NIR-conPK11195 uptake and TSPO-specificity were validated in TSPO-expressing human breast adenocarcinoma cells (MDA-MB-231). In vivo NIR-conPK11195 biodistribution and accumulation were quantitatively evaluated in athymic nude mice bearing MDA-MB-231 xenografts. RESULTS Fluorescence micrographs illustrated intracellular labeling of MDA-MB-231 cells by NIR-conPK11195. Quantitative uptake and competition assays demonstrated dose-dependent (p < 0.001) and TSPO-specific (p < 0.001) NIR-conPK11195 uptake. In vivo, NIR-conPK11195 preferentially labeled MDA-MB-231 tumors with an 11-fold (p < 0.001) and 7-fold (p < 0.001) contrast enhancement over normal tissue and unconjugated NIR dye, respectively. CONCLUSIONS NIR-conPK11195 appears to be a promising TSPO-targeted molecular imaging agent for visualization and quantification of breast cancer cells in vivo. This research represents the first study to demonstrate the feasibility of TSPO imaging as an alternative breast cancer imaging approach.
Collapse
|
17
|
Cappelli A, Mancini A, Sudati F, Valenti S, Anzini M, Belloli S, Moresco RM, Matarrese M, Vaghi M, Fabro A, Fazio F, Vomero S. Synthesis and biological characterization of novel 2-quinolinecarboxamide ligands of the peripheral benzodiazepine receptors bearing technetium-99m or rhenium. Bioconjug Chem 2008; 19:1143-53. [PMID: 18510350 DOI: 10.1021/bc700437g] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Potential receptor imaging agents based on Tc-99m for the in vivo visualization of the peripheral benzodiazepine receptor (PBR) have been designed on the basis of the information provided by the previously published structure-affinity relationship studies, which suggested the existence of tolerance to voluminous substituents in the receptor area interacting with 3-position of the quinoline nucleus of 2-quinolinecarboxamides 5. In the first step of the investigation, the stereoelectronic features of the above-indicated receptor area were also probed by means of 4-phenyl-3-[(1-piperazinyl)methyl]-2-quinolinecarboxamide derivatives bearing different substituents on the terminal piperazine nitrogen atom (compounds 6a-f). The structure-affinity relationship data confirmed the existence of a tolerance to bulky lipophilic substituents and stimulated the design of bifunctional ligands based on the 4-phenyl-3-[(1-piperazinyl)methyl]-2-quinolinecarboxamide moiety (compounds 6h,j,k,m). The submicromolar PBR affinity of rhenium complexes 6j,m suggests that the presence of their metal-ligand moieties with encaged rhenium is fairly compatible with the interaction with the PBR binding site. Thus, in order to obtain information on the in vivo behavior of these bifunctional ligands, (99m)Tc-labeled compounds 6h,k were synthesized and evaluated in preliminary biodistribution and single photon emission tomography (SPET) studies. The results suggest that both tracers do not present a clear preferential distribution in tissues rich in PBR, probably because of their molecular dimensions, which may hamper both the intracellular diffusion toward PBR and the interaction with the binding site.
Collapse
Affiliation(s)
- Andrea Cappelli
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cappelli A, Matarrese M, Moresco RM, Valenti S, Anzini M, Vomero S, Turolla EA, Belloli S, Simonelli P, Filannino MA, Lecchi M, Fazio F. Synthesis, labeling, and biological evaluation of halogenated 2-quinolinecarboxamides as potential radioligands for the visualization of peripheral benzodiazepine receptors. Bioorg Med Chem 2006; 14:4055-66. [PMID: 16495062 DOI: 10.1016/j.bmc.2006.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 02/01/2006] [Accepted: 02/03/2006] [Indexed: 11/30/2022]
Abstract
The previous exploration of the structure-affinity relationships concerning 4-phenyl-2-quinolinecarboxamide peripheral benzodiazepine receptor (PBR) ligands 6 showed as an interesting result the importance of the presence of a chlorine atom in the methylene carbon at position 3 of the quinoline nucleus. The subnanomolar PBR affinity shown by N-benzyl-3-chloromethyl-N-methyl-4-phenyl-2-quinolinecarboxamide (6b) suggested its chlorine atom to be replaced with other halogens in order to optimize the interaction of the quinolinecarboxamide derivatives with PBR and to develop suitable candidates for positron emission tomography (PET) or single photon emission computed tomography (SPECT) studies. The binding studies led to the discovery of fluoromethyl derivative 6a, which showed an IC50 value of 0.11 nM and is, therefore, one of the most potent PBR ligands so far described. Fluoromethyl derivative 6a has been labeled with 11C (t1/2=20.4 min, beta+=99.8%) starting from the corresponding des-methyl precursor (14) using [11C]CH3I in the presence of tetrabutylammonium hydroxide in DMF with a 35-40% radiochemical yield (corrected for decay) and 1.5 Ci/micromol of specific radioactivity. Ex vivo rat biodistribution and inhibition (following intravenous pre-administration of PK11195) studies showed that [11C]6a rapidly and specifically accumulated in PBR-rich tissues such as heart, lung, kidney, spleen, and adrenal, and at a lower level in other peripheral organs and in the brain. The images obtained in mouse with small animal YAP-(S)PET essentially confirmed the result of the ex vivo biodistribution experiments. The biological data suggest that [11C]6a is a promising radioligand for peripheral benzodiazepine receptor PET imaging in vivo.
Collapse
Affiliation(s)
- Andrea Cappelli
- Dipartimento Farmaco Chimico Tecnologico and European Research Centre for Drug Discovery and Development, Università degli Studi di Siena, Via A. Moro, 53100 Siena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Homes TP, Mattner F, Keller PA, Katsifis A. Synthesis and in vitro binding of N,N-dialkyl-2-phenylindol-3-yl-glyoxylamides for the peripheral benzodiazepine binding sites. Bioorg Med Chem 2006; 14:3938-46. [PMID: 16480880 DOI: 10.1016/j.bmc.2006.01.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 01/16/2006] [Accepted: 01/20/2006] [Indexed: 11/16/2022]
Abstract
A series of N,N-dialkyl-2-phenylindol-3-ylglyoxylamides bearing the halogens iodine and bromine were synthesised and their binding affinity for the peripheral benzodiazepine binding sites (PBBS) in rat kidney mitochondrial membranes was evaluated using [(3)H]PK11195. Central benzodiazepine receptor (CBR) affinities were also evaluated in rat cortices using [(3)H]flumazenil to determine their selectivity for PBBS over CBR. The tested compounds had PBBS binding affinities (IC(50)) ranging from 7.86 to 618 nM, with all compounds showing high selectivity over the CBR (CBR IC(50) > 5000 nM). Among the 12 compounds tested, those with a diethylamide group were the most potent. The highest affinity iodinated PBBS ligand, N,N-diethyl-[5-chloro-2-(4-iodophenyl)indol-3-yl]glyoxylamide, was radiolabelled with iodine-123. This high affinity and selective radioligand may be useful for imaging neurodegeneration, inflammation and tumours using single photon emission computed tomography.
Collapse
Affiliation(s)
- Taryn P Homes
- Department of Chemistry, University of Wollongong, NSW 2500, Wollongong, Australia
| | | | | | | |
Collapse
|
20
|
Giusti L, Costa B, Viacava P, Castagna M, Iacconi P, Ricci RE, Zaccagnini M, Miccoli P, Lucacchini A. Peripheral type benzodiazepine receptor in human parathyroid glands: up-regulation in adenoma. J Endocrinol Invest 2004; 27:826-31. [PMID: 15648546 DOI: 10.1007/bf03346276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this study we report the presence of peripheral benzodiazepine receptors (PBRs) in human parathyroid glands and describe the effect of their benzodiazepine type ligands on parathyroid cell function. PBR binding features in normal parathyroid tissue were characterized and compared to parathyroid adenoma, using a specific and selective ligand for PBR, [3H] 1-(2-chlorophenyl)-N-methyl-N-(1-methyl-propyl)-3-isoquinoline-carboxamide ([3H]PK11195). Affinity and density of [3H]PK11195 binding sites in homogenate membrane preparations from adenomatous and normal tissues were determined. Parathyroid adenoma showed a statistically significant 2.2 fold increase of [3H]PK11195 binding sites, while the affinity remained unchanged. Our results represent the first evidence of PBRs in parathyroid glands and suggest for them a role in influencing PTH release. A clear trend of PBR up-regulation in parathyroid adenoma was also found.
Collapse
Affiliation(s)
- L Giusti
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Chelli B, Lena A, Vanacore R, Da Pozzo E, Costa B, Rossi L, Salvetti A, Scatena F, Ceruti S, Abbracchio MP, Gremigni V, Martini C. Peripheral benzodiazepine receptor ligands: mitochondrial transmembrane potential depolarization and apoptosis induction in rat C6 glioma cells. Biochem Pharmacol 2004; 68:125-34. [PMID: 15183124 DOI: 10.1016/j.bcp.2004.03.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2003] [Accepted: 03/05/2004] [Indexed: 10/26/2022]
Abstract
The peripheral benzodiazepine receptor (PBR) is a component of a multiprotein complex, located at the contact site between the inner and outer mitochondrial membranes, which constitutes the mitochondrial permeability transition (MPT)-pore. The opening of the MPT-pore, leading to the transmembrane mitochondrial potential (DeltaPsi(m)) dissipation, is a critical event in the mechanism of apoptosis. In the present work, we investigated the ability of the specific PBR ligands, PK 11195 or Ro5-4864, to affect mitochondrial potential and to induce apoptotic cell death in rat C6 glioma cells. Both specific ligands inhibited cell survival in a dose- and time-dependent manner, as assessed by MTS conversion assay, whereas the non-site selective ligand Diazepam or the low-affinity benzodiazepine Clonazepam showed no significant effects. After cell exposure to PK 11195 or Ro5-4864 we evidenced typical alterations of apoptotic cell death such as DNA fragmentation and chromatin condensation assessed by flow cytometric and transmission electron microscopy (TEM) analysis, respectively. Activation of the "effector" caspase-3 confirmed the ability of specific PBR ligands to induce apoptosis. Moreover, PK 11195 and Ro5-4864 induced a decrease of DeltaPsi(m), as evidenced by JC-1 flow cytometry analysis. Our data demonstrate the pro-apoptotic effects of specific PBR ligands on rat C6 glioma cells.
Collapse
Affiliation(s)
- Beatrice Chelli
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
As photodynamic therapy (PDT) becomes established as a treatment for cancer, there is increasing interest in identifying critical mechanisms of cell killing and understanding the bases for effective photosensitizers. The existence of multiple cellular targets makes it difficult to distinguish the critical events leading to cell death from PDT. However, with more sensitive techniques to detect photosensitizer localization, the isolation of PDT-resistant and -sensitive mutants and the use of innovative molecular and biochemical strategies to map cellular events occurring during and after photosensitization, some order is emerging from the chaos. The subcellular localization of many photosensitizers and the early responses to light activation indicate that mitochondria play a major role in photodynamic cell death. PDT with many agents which damage or inhibit different or multiple mitochondrial targets has many of the desirable characteristics for an effective anti-cancer therapy.
Collapse
Affiliation(s)
- J Morgan
- Department of Dermatology, MRC2, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | |
Collapse
|
23
|
Lavicka J, Sarisský M, Mirossay A, Sulla I, Mojzis J, Mirossay L. Diazepam enhances etoposide-induced cytotoxicity in U-87 MG human glioma cell line. Fundam Clin Pharmacol 2001; 15:201-7. [PMID: 11468031 DOI: 10.1046/j.1472-8206.2001.00030.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Various approaches might be employed in an effort to increase efficacy of the chemotherapeutic treatment of cancer. Recently, various modulators of anticancer therapy effectiveness have been studied. Antiproliferative effects of peripheral benzodiazepine receptor (PBR) ligands might be exploited to enhance cytotoxic effect of a chemotherapeutic drug towards cancer cells. In this work, we sought to enhance cytotoxic effect of etoposide (VP-16) by a PBR ligand, diazepam (DZ) in U-87 MG human glioma cells. Cytotoxicity of VP-16, DZ and their combinations was assessed by using the microculture MTT assay. Cell survival, effective concentrations (EC) and the onset of cytotoxic effect were determined. After 72 h of cultivation, survival of U-87 MG cells was reduced to 57 +/- 7% in the presence of VP-16 at 12.5 microg/mL alone, whereas DZ at 10-4 mol/L alone caused 28 +/- 6% reduction in cell survival. Coincubation of VP-16 at 12.5 microg/mL with DZ at 10-4 mol/L led to a further decrease in cell survival to 45 +/- 6%. Furthermore, DZ at 10-4 mol/L significantly decreased effective concentrations, EC10, EC30 and EC50, of VP-16 and the dose-response curves were shifted to the left. Addition of DZ at 10-4 mol/L to VP-16 also facilitated the onset of its cytotoxic effect. The same decrease in survival was thus achieved approximately 30 h earlier in comparison with VP-16 alone. However, DZ at 10-9 mol/L failed both to exert any effect on glioma cells survival and enhance cytotoxic effect of VP-16. DZ at 10-4 mol/L was capable of both reducing U-87 MG glioma cells survival when applied alone and also enhancing the cytotoxic effect of VP-16. No such observation was made for the lower concentrations of DZ. Potential implementation of diazepam in the antiglioma/anticancer armamentarium awaits further experimentation but phase I and phase II clinical trials could be suggested.
Collapse
Affiliation(s)
- J Lavicka
- Department of Pharmacology, Faculty of Medicine, Safárik University, Kosice, Slovakia
| | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Venturini I, Alho H, Podkletnova I, Corsi L, Rybnikova E, Pellicci R, Baraldi M, Pelto-Huikko M, Helén P, Zeneroli ML. Increased expression of peripheral benzodiazepine receptors and diazepam binding inhibitor in human tumors sited in the liver. Life Sci 1999; 65:2223-31. [PMID: 10576594 DOI: 10.1016/s0024-3205(99)00487-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The peripheral benzodiazepine receptor system triggers intracellular metabolic events and has been associated with cell proliferation. Its endogenous ligand, the diazepam binding inhibitor, contributes to steroidogenesis by promoting cholesterol delivery to the inner mitochondrial membrane. The present study was undertaken to verify whether this system is altered in tumors sited in the liver. Peripheral benzodiazepine receptors and diazepam binding inhibitor were studied using immunocytochemistry and in situ hybridization in 9 human tumors sited in the liver, in liver hyperplasia, cirrhotic nodular regeneration, intestinal adenocarcinoma and in surrounding non-tumoral tissue. Immunocytochemical staining and in situ hybridization demonstrated that peripheral benzodiazepine receptors and diazepam binding inhibitor were more prominently expressed in neoplastic cells than in non-tumoral tissue. They were present in the same cells, suggesting that diazepam binding inhibitor may act in an intracrine manner in these cells. Higher peripheral benzodiazepine receptors and diazepam binding inhibitor expression in tumor cells suggest an implication of this system in the metabolism of neoplastic cells. Furthermore the evaluation of peripheral benzodiazepine receptor and diazepam binding inhibitor expression might be useful in evaluating malignancy and in diagnostic approaches of tumors in liver tissue.
Collapse
Affiliation(s)
- I Venturini
- Cattedra di Semeiotica e Metodologia Medica, Università di Modena, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Derlon JM. The in vivo metabolic investigation of brain gliomas with positron emission tomography. Adv Tech Stand Neurosurg 1999; 24:41-76. [PMID: 10050211 DOI: 10.1007/978-3-7091-6504-1_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- J M Derlon
- Service de Neurochirurgie, CHU, Caen, France
| |
Collapse
|
27
|
Venturini I, Zeneroli ML, Corsi L, Avallone R, Farina F, Alho H, Baraldi C, Ferrarese C, Pecora N, Frigo M, Ardizzone G, Arrigo A, Pellicci R, Baraldi M. Up-regulation of peripheral benzodiazepine receptor system in hepatocellular carcinoma. Life Sci 1998; 63:1269-80. [PMID: 9771915 DOI: 10.1016/s0024-3205(98)00388-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increased number of peripheral benzodiazepine receptors (PBRs) have been found in some tumors outside the liver. The present study was to verify whether the PBR system is altered in hepatocellular carcinoma (HCC). The levels of endogenous benzodiazepine-like compounds (BZDs), measured by radioreceptor binding technique after HPLC purification and the endogenous ligand for PBRs, termed diazepam binding inhibitor (DBI), measured by radioimmunoassay utilizing a specific antibody for human DBI, were studied in the blood of 15 normal subjects, 12 liver cirrhosis and 10 patients with HCC. The levels of BZDs in serum were increased hundred fold in liver cirrhosis patients and slightly elevated in HCC patients. DBI was found to be increased in HCC patients. The binding recognition sites for PBRs (Bmax) were increased 4 to 7 fold in HCC tissue in comparison with that found in non-tumoral liver tissue (NTLT). On the contrary the concentrations of DBI were found to be significantly decreased in HCC tissue in comparison with the respective NTLT. These results seem to suggest an implication of PBRs and of their putative endogenous ligands in the metabolism of these neoplastic cells and possibly in their proliferation. The up-regulation of PBRs found in HCC tissue seems to indicate an increased functional activity of these receptors and opens up the possibility of new pharmacological and diagnostic approaches while the changes in the circulating endogenous ligands for the above receptors might be envisaged as early markers of tumorigenesis in liver cirrhosis.
Collapse
Affiliation(s)
- I Venturini
- Cattedra di Semeiotica e Metodologia Medica, Università di Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Miyazawa N, Hamel E, Diksic M. Assessment of the peripheral benzodiazepine receptors in human gliomas by two methods. J Neurooncol 1998; 38:19-26. [PMID: 9540054 DOI: 10.1023/a:1005933226966] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study was designed to evaluate the density of peripheral benzodiazepine receptor (PBR) sites as a function of tumor malignancy in human gliomas, and to compare the results obtained with autoradiographic and liquid scintillation measurements performed on the same tissue specimens. In vitro binding of [3H]PK-11195[1-(2-chlorophenyl)-N-methyl-(1-methylpropyl)-3-isoguinol ine carboxamide] to human gliomas in radioligand binding studies revealed a significantly higher level (about 3 fold) of PBR binding sites in both low grade and high grade gliomas as compared to normal cortex. The Bmax (mean +/- SD) of high and low grade gliomas, when entire tissue sections were measured by autoradiography, was 5.5 +/- 0.3 pmol/mg-tissue (n = 5) and 1.8 +/- 0.9 pmol/mg-tissue (n = 6), respectively, although it was evident that there was area of hot spots in the high grade tumors. This difference was significant (p < 0.05; two-tailed t-test). Similarly, the KD values (dissociation constant; nM) between the high (KD = 20.4 +/- 1.3 nM) and low (KD = 14.3 +/- 2.1 nM) grade gliomas were significantly different. A significant difference in binding site density (Bmax) between the two types of gliomas was also obtained in liquid scintillation measurements. The hot spot areas which showed the most intense binding of [3H]PK-11195 had KD of 24.5 +/- 1.0 nM and Bmax of 6.2 +/- 0.42 pmol/mg-tissue, values significantly higher (p < 0.05, two-tailed t-test) than those obtained when the entire tissue section was measured. The data on the Bmax/KD ratios presented here suggest that it might be possible to differentiate high from low grade gliomas in human by in vivo imaging with 11C-labelled PK-11195.
Collapse
Affiliation(s)
- N Miyazawa
- Cone Laboratory for Neurosurgical Research, Montreal, Canada
| | | | | |
Collapse
|
29
|
Takeda N, Diksic M, Yamamoto YL. The sequential changes in DNA synthesis, glucose utilization, protein synthesis, and peripheral benzodiazepine receptor density in C6 brain tumors after chemotherapy to predict the response of tumors to chemotherapy. Cancer 1996; 77:1167-79. [PMID: 8635140 DOI: 10.1002/(sici)1097-0142(19960315)77:6<1167::aid-cncr25>3.0.co;2-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Monitoring therapy in patients with brain tumors is very difficult and unreliable. It has been shown that there is no good correlation between tumor sensitivity measured in vitro and in situ tumor response to therapies. METHODS Sequential changes in tumor size, number of DNA synthesizing cells (labelling index [LI]), glucose utilization (LCGU), protein synthesis (LCPS), and peripheral benzodiazepine receptor (PBR) density were examined after chemotherapy for seven days. This was done using antibromodeoxyuridine immunohistochemical stain and multiple tracer quantitative autoradiography in a C6 rat brain with an implanted glioma. On Day 10 after inoculation, the rats were divided into 5 experimental groups: (1) a nontreatment group (control Group 1); (2) a group received 5% dextrose intraarterial (IA) administration (control Group 2); (3) a group received 1,3-bis-(2-chloroethyl) nitrosourea (BCNU) intravenous (i.v.) administration (Group 3) (5% dextrose was solvent); (4) a group received BCNU IA administration (Group 4) (5% dextrose was solvent); and (5) a group received sarcosinamide chloroethyl nitrosourea (SarCNU) IA administration (Group 5) (solvent as for the BCNU group). RESULTS Three treatments showed a significant decrease (P < 0.003) in tumor growth. The most effective treatment was BCNU IA and SarCNU IA was moderately effective. BCNU i.v. showed no effect on tumor growth when compared with the two control groups. The change in the peak LI correlated well with the peak LCGU. These parameters decreased markedly and significantly in both Group 4 and Group 5 from Day 1 after treatment. The rates of the decrease in these biologic factors also correlated well with a decrease in the tumor growth. The LCPS did not correlate with a decrease in the LI or LCGU. The dissociation constant (Kd) and densities of the receptors PBR (B max) did not change significantly in any of the treatment groups during the observation period. CONCLUSIONS From the results presented, we concluded that changes in the LI and LCGU represent the most reliable parameters with which to predict the response or sensitivity of this glial tumor to the treatments applied. These data suggest that if changes in peak LCGU were measured in tumors using positron emission tomography, they might be instrumental in providing in vivo information about the sensitivity of a tumor to a given treatment without the need for repeated tumor biopsy.
Collapse
Affiliation(s)
- N Takeda
- Cone Laboratory for Neurosurgical Research, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
30
|
Miyazawa N, Diksic M, Yamamoto Y. Chronological study of peripheral benzodiazepine binding sites in the rat brain stab wounds using [3H] PK-11195 as a marker for gliosis. Acta Neurochir (Wien) 1995; 137:207-16. [PMID: 8789663 DOI: 10.1007/bf02187195] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Chronological studies of the development of the peripheral benzodiazepine receptor sites were undertaken with the goal of evaluating the sensitivity of this marker for the study of the gliosis development in the injured brain. No significant increase in [3H] PK-11195 binding occurred in the rat brain stab wound one day following the puncture. A significant increase in the receptor density (Bmax) from the second day onward was observed. The Bmax reached its highest levels in the grey matter on the sixth day after a 23-gauge needle wound (8.75 +/- 0.09; pmol mg-tissue-1) and on the seventh day after an 18-gauge needle wound (8.98 +/- 0.31 pmol mg-tissue-1). In the white matter, the Bmax was greatest seven days after the wound (3.42 +/- 0.07; pmol mg-tissue-1; 23-gauge needle and 3.56 +/- 0.1 pmol mg-tissue-1 in the 18-gauge needle injury). Between 30 and 60 days after the wound, the Bmax was significantly lower than the Bmax observed between 6 and 14 days. The Bmax in the wound produced with needles was seven to eight times greater than the Bmax in the grey matter of the ipsilateral and contralateral cortices. Histological examination showed that there were no astrocytes or macrophages in the stab wound one day after the lesion. However, the glial fibrillary acidic protein positive cells and macrophages appeared on D3 after an injury. Gliosis, as measured by the PK-11195 binding, was also observed in the remote contralateral cortex. Data shows that PK-11195 binding is a very sensitive method of evaluating brain injury and could be of great value in studying progressive injuries in the living human brain in conjunction with positron emission tomography.
Collapse
Affiliation(s)
- N Miyazawa
- Cone Laboratory for Neurosurgical Research, Montreal Neurological Institute, Quebec, Canada
| | | | | |
Collapse
|
31
|
Chen TC, Rabb C, Apuzzo ML. Complex Technical Methodologies and Their Applications in the Surgery of Intracranial Meningiomas. Neurosurg Clin N Am 1994. [DOI: 10.1016/s1042-3680(18)30530-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Ferrarese C, Pierpaoli C, Linfante I, Bobo RH, Guthrie B, Kufta C, Duhaney MO, Melisi J, Fulham MJ. Peripheral benzodiazepine receptors and glucose metabolism in human gliomas. J Neurooncol 1994; 22:15-22. [PMID: 7714547 DOI: 10.1007/bf01058351] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Peripheral benzodiazepine receptors (PBR) are increased in gliomas and augmented glucose metabolism is seen in malignant brain tumors. We investigated the relationship between PBR density (Bmax) and glucose utilization rate (GUR) in 17 patients with cerebral gliomas of different grades. PBR Bmax was assessed by [3H]PK-11195 in vitro binding in surgical specimens and GUR was measured by Positron Emission Tomography with [18F]2-Fluorodeoxyglucose before the surgery. In untreated tumors there was a positive correlation between PBR Bmax and GUR (2r = 0.84). This correlation was not observed in patients who had been treated with radiation and/or chemotherapy prior to surgery (r2 = 0.13). In addition, in untreated patients, the increase in PBR density and GUR appeared to be related to the degree of malignancy.
Collapse
Affiliation(s)
- C Ferrarese
- Department of Neurosurgery, George Washington University Medical Center, Washington DC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Pike VW, Halldin C, Crouzel C, Barré L, Nutt DJ, Osman S, Shah F, Turton DR, Waters SL. Radioligands for PET studies of central benzodiazepine receptors and PK (peripheral benzodiazepine) binding sites--current status. Nucl Med Biol 1993; 20:503-25. [PMID: 8389223 DOI: 10.1016/0969-8051(93)90082-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The status of the radiochemical development and biological evaluation of radioligands for PET studies of central benzodiazepine (BZ) receptors and the so-called peripheral benzodiazepine binding sites, here discriminated and referred to as PK binding sites, is reviewed against current pharmacological knowledge, indicating those agents with present value and those with future potential. Practical recommendations are given for the preparation of two useful radioligands for PET studies, [N-methyl-11C]flumazenil for central BZ receptors, and [N-methyl-11C]PK 11195 for PK binding sites. Quality assurance and plasma metabolite analysis are also reviewed for these radioligands and practical recommendations are given on methodology for their performance.
Collapse
Affiliation(s)
- V W Pike
- MRC Cyclotron Unit, Hammersmith Hospital, London, England
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hawkins RA, Hoh C, Glaspy J, Choi Y, Dahlbom M, Rege S, Messa C, Nietszche E, Hoffman E, Seeger L. The role of positron emission tomography in oncology and other whole-body applications. Semin Nucl Med 1992; 22:268-84. [PMID: 1439872 DOI: 10.1016/s0001-2998(05)80121-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Imaging and quantifying biochemical and physiological processes with PET clearly has major potential significance for all organ systems and many disease states. Although the full utility and potential of emerging new applications of PET in organs other than the heart and brain must be demonstrated in basic and clinical research studies, the rapidly accumulating aggregate experience in oncology in particular, and in other organ systems and disease states as well, indicates that PET is now truly becoming a modality of both clinical and investigative use for the body as a whole as well as for specific organ systems. Whole-body PET FDG imaging (Fig 9) illustrates the potential of biochemical imaging to map the distribution of cancer throughout the body. With the growing list of radiopharmaceutical and quantitative techniques applicable to cancer studies with PET, this field will continue to realize significant growth.
Collapse
Affiliation(s)
- R A Hawkins
- Department of Radiological Sciences, UCLA School of Medicine 90024
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Takada A, Mitsuka S, Diksic M, Yamamoto YL. Autoradiographic study of peripheral benzodiazepine receptors in animal brain tumor models and human gliomas. Eur J Pharmacol 1992; 228:131-9. [PMID: 1332878 DOI: 10.1016/0926-6917(92)90022-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In vitro binding of [3H]PK-11195 (1-(2-chlorophenyl)-N-methyl-(1- methylpropyl)-3-isoquinoline carboxamide) in rodent AA ascites and C6 glioma as well as in human gliomas was investigated. The Bmax (mean +/- S.D.) of AA ascites tumor and C6 glioma is 1.39 +/- 0.15 pmol/mg tissue and 4.50 +/- 0.76 pmol/mg tissue, respectively. This Bmax is 9 and 30 times, respectively, higher than the one found in the rat cortex (0.15 +/- 0.03 pmol/mg tissue). A Bmax of 1.26 +/- 0.24 pmol/mg tissue and 0.64 +/- 0.08 pmol/mg tissue was found in human malignant and low grade gliomas respectively. This Bmax value should be compared to 0.35 +/- 0.04 pmol/mg tissue found in the normal human cortex. There are significant (P less than 0.05) differences between Bmax in tumors and normal cortex. There was no significant difference in KD between the malignant and low grade gliomas. C6 glioma has a KD significantly greater than rat cortex. In some cases of human low grade gliomas, kinetic measurements suggested the presence of two affinity receptor sites. However, at this time, heterogeneity of the tissue cannot be excluded as being at least in part a source of this.
Collapse
Affiliation(s)
- A Takada
- Cone Laboratory for Neurosurgical Research, Montreal Neurological Institute, Canada
| | | | | | | |
Collapse
|
36
|
Shiraishi T, Black KL, Ikezaki K, Becker DP. Peripheral benzodiazepine induces morphological changes and proliferation of mitochondria in glioma cells. J Neurosci Res 1991; 30:463-74. [PMID: 1666128 DOI: 10.1002/jnr.490300303] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Peripheral benzodiazepine (PBD) receptors are localized on the mitochondrial membrane and are highly expressed in brain tumors compared to normal brain. To elucidate the biological role of the PBD receptor on mitochondria, we examined the effect of PBDs on mitochondrial morphology in C6 and T98G glioma cells using rhodamine 123 and quantitative electron microscopy. In cells incubated in serum-free medium alone, mitochondria were distributed in a filamentous pattern throughout the cytoplasm. By contrast, the mitochondria aggregated in the perinuclear region in PK11195 or Ro5-4864 (10 nM) treated cells. Quantitative electron micrography revealed a 250% increased in the number of mitochondria with elongated cristae and a fivefold increase in dividing mitochondria in PK11195-treated cells compared with cells incubated in serum-free medium alone. PBD treatment also resulted in vacuolation within the matrix and mitochondrial swelling. These data suggest that PBDs influence mitochondrial morphology and induce mitochondrial replication in cultured glioma cells.
Collapse
Affiliation(s)
- T Shiraishi
- Brain Research Institute, Jonsson Cancer Center, Los Angeles, California
| | | | | | | |
Collapse
|
37
|
Diorio D, Welner SA, Butterworth RF, Meaney MJ, Suranyi-Cadotte BE. Peripheral benzodiazepine binding sites in Alzheimer's disease frontal and temporal cortex. Neurobiol Aging 1991; 12:255-8. [PMID: 1652108 DOI: 10.1016/0197-4580(91)90106-t] [Citation(s) in RCA: 93] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Much evidence has accumulated to suggest that the peripheral type benzodiazepine (PBZ) binding site has a predominantly glial localization. Elevated PBZ binding densities have been reported in various models of brain damage, apparently reflecting glial proliferation in response to neurodegeneration. In the present study, PBZ receptor densities were examined in frontal and temporal cortex of Alzheimer's disease (AD) patients using the ligand [3H]PK 11195. There was a highly significant (p less than 0.01) increase in PBZ binding sites in the temporal cortex from AD patients. In the frontal cortex, a moderate increase was observed that approached statistical significance (p = 0.07). Decreased choline acetyltransferase activity was observed in both regions. These findings offer support for the potential use of the PBZ binding site as a marker for gliosis associated with neuronal cell death.
Collapse
Affiliation(s)
- D Diorio
- Department of Psychiatry, Douglas Hospital Research Centre, McGill University, Verdun, Quebec, Canada
| | | | | | | | | |
Collapse
|
38
|
Abstract
Probing the regional distribution and affinity of receptors in the brain, in vivo, in human and non human primates has become possible with the use of selective ligands labelled with positron emitting radionuclides and positron emission tomography (PET). After describing the techniques used in positron emission tomography to characterize a ligand receptor binding and discussing the choice of the label and the limitations and complexities of the in vivo approach, the results obtained in the PET studies of various neurotransmission systems: dopaminergic, opiate, benzodiazepine, serotonin and cholinergic systems are reviewed.
Collapse
Affiliation(s)
- B Mazière
- Service Hospitalier Frédéric Joliot, Commissariat à l'Energie Atomique, Orsay, France
| | | |
Collapse
|
39
|
Ikezaki K, Black KL, Toga AW, Santori EM, Becker DP, Smith ML. Imaging peripheral benzodiazepine receptors in brain tumors in rats: in vitro binding characteristics. J Cereb Blood Flow Metab 1990; 10:580-7. [PMID: 2161415 DOI: 10.1038/jcbfm.1990.101] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Peripheral benzodiazepine binding constants for transplanted RG-2 gliomas and HD and LK Walker 256 tumors (metastatic breast carcinoma) were determined in Wistar rats using autoradiography. In addition, Kd and Bmax parameters for peripheral benzodiazepine receptors on RG-2 tumors were directly visualized using digital image analysis of autoradiograms. High specific binding of [3H]PK11195, a selective peripheral benzodiazepine ligand, had excellent topographical correlation to areas of histologically verified tumor. Scatchard analysis suggested a single class of peripheral binding sites with similar binding affinities in RG-2 and LK Walker 256 tumors and normal cortex. Bmax was 20-fold greater in glial tumors and 11.6- and 10.6-fold greater in LK and HK Walker 256 tumors, respectively, compared to normal cortex. The location of metastatic tumors, either intracerebrally or subcutaneously, did not effect their Kd or Bmax values. Kd and Bmax values for RG-2 tumors were similar whether determined densitometrically or by direct visualization with image analysis. Binding parameters within normal brain were difficult to visualize by image analysis due to the low level of specific binding. The ability to label specifically intracerebral tumor cells and to characterize the binding parameters shown in this study suggest that peripheral benzodiazepine receptor ligands could be utilized by PET to analyze directly a variety of tumors in humans.
Collapse
Affiliation(s)
- K Ikezaki
- Division of Neurosurgery, University of California, Los Angeles, School of Medicine 90024
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The effects of peripheral benzodiazepine receptor ligands on cell proliferation were evaluated. PK11195 increased the growth rate of C6 glioma cells by 20-30% in the nanomolar range in serum free medium. [3H]thymidine incorporation into C6 glioma cells also were increased 22% and 25% after treatment by PK11195 and Ro5-4864, respectively. The effect of PK11195 as a mitogenic agent was estimated by mitogenic agent was estimated by [3H]thymidine incorporation using Swiss 3T3 cells. PK11195 increased DNA synthesis 170% over control at 10 nM. Higher concentrations of benzodiazepines showed inhibition of the DNA synthesis. Peripheral benzodiazepine binding sites underwent downregulation after exposure to serum free medium or to 10 nM PK11195. These findings suggest that peripheral benzodiazepines may be involved in the regulation of cell proliferation as a growth factor in lower concentration and as a antiproliferative agent in higher concentration.
Collapse
Affiliation(s)
- K Ikezaki
- Jonsson Cancer Center, Division of Neurosurgery, UCLA School of Medicine 90024
| | | |
Collapse
|
41
|
Bockhorst K, Höhn-Berlage M, Kocher M, Hossmann KA. Proton relaxation enhancement in experimental brain tumors--in vivo NMR study of manganese(III)TPPS in rat brain gliomas. Magn Reson Imaging 1990; 8:499-504. [PMID: 2392036 DOI: 10.1016/0730-725x(90)90058-a] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The effect of manganese(III)tetraphenylporphine sulfonate (MnTPPS) on the relaxation enhancement of NMR images (MRI) was studied in experimental brain tumors in rats. Brains were inoculated with the glioma cell line F98 12 to 19 days before the NMR experiment, and the effect of MnTPPS (0.25 mmol/kg body weight) was investigated 2 and 4 days after intraperitoneal injection. After MnTPPS addition tumors could be clearly distinguished by the brightness from the surrounding brain whereas they were barely visible without contrast enhancement. At SE time of 25 msec and TR time of 3500 msec the ratio of magnetization values of tumor versus normal grey matter increased from 0.98 +/- 0.08 to 1.24 +/- 0.09 (means +/- SD). When TR was shortened to 1100 msec contrast enhancement further increased to 1.77 +/- 0.25. These results demonstrate for the first time that MnTPPS is an efficient agent for contrast enhancement of brain tumors.
Collapse
Affiliation(s)
- K Bockhorst
- Max-Planck-Institut für neurologische Forschung, Abteilung für experimentelle Neurologie, Cologne, FRG
| | | | | | | |
Collapse
|
42
|
Mazière B, Mazière M. Where have we got to with neuroreceptor mapping of the human brain? EUROPEAN JOURNAL OF NUCLEAR MEDICINE 1990; 16:817-35. [PMID: 2170141 DOI: 10.1007/bf00833018] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the past two decades, tritiated radioligand receptor binding, a tool commonly used to investigate the site of action of drugs in laboratory animals, has provided a vast body of information on neuropharmacology and neurobiology. Several neurological and psychiatric diseases have been related to neurotransmitter and receptor disorders. In order to study ligand interactions with receptors in vivo in humans, new tracers capable of carrying a gamma-emitting radionuclide to the receptor have been designed. Emission computerized tomography (ECT) techniques such as positron (PET) or single photon emission tomography (SPET) allow monitoring of the time-course of regional tissue concentration of these radiolabelled ligands. PET and SPET each have their inherent advantages and drawbacks. The cyclotron-based technology of PET is a demanding and expensive technique that, to date, is still mainly reserved for research purposes. It is hoped that once the scientific basis of a physiopathological study is established using PET, diagnostic information might be provided by the more readily available SPET technology. The purpose of this article is to review the current state of receptor-binding gamma-emitting radioligands and to present the clinical potential of these new kinds of radiopharmaceuticals in clinical investigation.
Collapse
Affiliation(s)
- B Mazière
- Service Hospitalier Frédéric Joliot Commissariat à l'Energie Atomique, Orsay, France
| | | |
Collapse
|