1
|
Soyer A, Goutal S, Leterrier S, Marie S, Larrat B, Selingue E, Winkeler A, Sarazin M, Bottlaender M, Tournier N. [ 18F]2-fluoro-2-deoxy-sorbitol ([ 18F]FDS) PET imaging repurposed for quantitative estimation of blood-brain barrier permeability in a rat model of Alzheimer's disease. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:822-829. [PMID: 38657857 DOI: 10.1016/j.pharma.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/05/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Numerous studies suggest that blood-brain barrier (BBB) dysfunction may contribute to the progression of Alzheimer's disease (AD). Clinically available neuroimaging methods are needed for quantitative "scoring" of BBB permeability in AD patients. [18F]2-fluoro-2-deoxy-sorbitol ([18F]FDS), which can be easily obtained from simple chemical reduction of commercial [18F]2-fluoro-2-deoxy-glucose ([18F]FDG), was investigated as a small-molecule marker of BBB permeability, in a pre-clinical model of AD using in vivo PET imaging. Chemical reduction of [18F]FDG to [18F]FDS was obtained with a 100% conversion yield. Dynamic PET acquisitions were performed in the APP/PS1 rat model of AD (TgF344-AD, n=3) compared with age-matched littermates (WT, n=4). The brain uptake of [18F]FDS was determined in selected brain regions, delineated from a coregistered rat brain template. The brain uptake of [18F]FDS in the brain regions of AD rats versus WT rats was compared using a 2-way ANOVA. The uptake of [18F]FDS was significantly higher in the whole brain of AD rats, as compared with WT rats (P<0.001), suggesting increased BBB permeability. Enhanced brain uptake of [18F]FDS in AD rats was significantly different across brain regions (P<0.001). Minimum difference was observed in the amygdala (+89.0±7.6%, P<0.001) and maximum difference was observed in the midbrain (+177.8±29.2%, P<0.001). [18F]FDS, initially proposed as radio-pharmaceutical to estimate renal filtration using PET imaging, can be repurposed for non-invasive and quantitative determination of BBB permeability in vivo. Making the best with the quantitative properties of PET imaging, it was possible to estimate the extent of enhanced BBB permeability in a rat model of AD.
Collapse
Affiliation(s)
- Amélie Soyer
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Sébastien Goutal
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Sarah Leterrier
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Solène Marie
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Benoit Larrat
- Centre d'études de Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Paris-Saclay University, 91191 Gif-sur-Yvette, France
| | - Erwan Selingue
- Centre d'études de Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Paris-Saclay University, 91191 Gif-sur-Yvette, France
| | - Alexandra Winkeler
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Marie Sarazin
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Michel Bottlaender
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Nicolas Tournier
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France.
| |
Collapse
|
2
|
Leterrier S, Goutal S, Hugon G, Goislard M, Saba W, Hosten B, Specklin S, Winkeler A, Tournier N. Imaging quantitative changes in blood-brain barrier permeability using [ 18F]2-fluoro-2-deoxy-sorbitol ([ 18F]FDS) PET in relation to glial cell recruitment in a mouse model of endotoxemia. J Cereb Blood Flow Metab 2024; 44:1117-1127. [PMID: 38441006 PMCID: PMC11179610 DOI: 10.1177/0271678x241236755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/06/2024]
Abstract
The quantitative relationship between the disruption of the blood-brain barrier (BBB) and the recruitment of glial cells was explored in a mouse model of endotoxemia. [18F]2-Fluoro-2-deoxy-sorbitol ([18F]FDS) PET imaging was used as a paracellular marker for quantitative monitoring of BBB permeability after i.v injection of increasing doses of lipopolysaccharide (LPS) or vehicle (saline, n = 5). The brain distribution of [18F]FDS (VT, mL.cm-3) was estimated using kinetic modeling. LPS dose-dependently increased the brain VT of [18F]FDS after injection of LPS 4 mg/kg (5.2 ± 2.4-fold, n = 4, p < 0.01) or 5 mg/kg (9.0 ± 9.1-fold, n = 4, p < 0.01) but not 3 mg/kg (p > 0.05, n = 7). In 12 individuals belonging to the different groups, changes in BBB permeability were compared with expression of markers of astrocyte (GFAP) and microglial cell (CD11b) using ex vivo immunohistochemistry. Increased expression of CD11b and GFAP expression was observed in mice injected with 3 mg/kg of LPS, which did not increase with higher LPS doses. Quantitative [18F]FDS PET imaging can capture different levels of BBB permeability in vivo. A biphasic effect was observed with the lowest dose of LPS that triggered neuroinflammation without disruptive changes in BBB permeability, and higher LPS doses that increased BBB permeability without additional recruitment of glial cells.
Collapse
Affiliation(s)
- Sarah Leterrier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Sébastien Goutal
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Gaëlle Hugon
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Maud Goislard
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Wadad Saba
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Benoit Hosten
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Simon Specklin
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Alexandra Winkeler
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| |
Collapse
|
3
|
Hosten B, Goutal S, Leterrier S, Corvo C, Breuil L, Barret O, Specklin S, Truillet C, Tournier N. Brain delivery enabled by transient blood-brain barrier disruption induced by regadenoson: a PET imaging study. Expert Opin Drug Deliv 2024; 21:797-807. [PMID: 38881261 DOI: 10.1080/17425247.2024.2369765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Regadenoson, an agonist of adenosine A2 receptors, enables transient blood-brain barrier (BBB) disruption. The relevance of regadenoson as a pharmacological strategy for brain delivery was investigated using in vivo PET imaging in rats. RESEARCH DESIGN AND METHODS Kinetic modeling of brain PET data was performed to estimate the impact of regadenoson (0.05 mg.kg-1, i.v.) on BBB permeation compared with control rats (n = 4-6 per group). Three radiolabeled compounds of different sizes, which do not cross the intact BBB, were tested. RESULTS Regadenoson significantly increased the BBB penetration (+116 ± 13%, p < 0.001) of [18F]2-deoxy-2-fluoro-D-sorbitol ([18F]FDS, MW = 183 Da), a small-molecule marker of BBB permeability. The magnitude of the effect was different across brain regions, with a maximum increase in the striatum. Recovery of BBB integrity was observed 30 min after regadenoson injection. Regadenoson also increased the brain penetration (+72 ± 45%, p < 0.05) of a radiolabeled nanoparticle [89Zr]AGuIX (MW = 9 kDa). However, the brain kinetics of a monoclonal antibody ([89Zr]mAb, MW = 150 kDa) remained unchanged (p > 0.05). CONCLUSIONS PET imaging showed the features and limitations of BBB disruption induced by regadenoson in terms of extent, regional distribution, and reversibility. Nevertheless, regadenoson enables the brain delivery of small molecules or nanoparticles in rats.
Collapse
Affiliation(s)
- Benoit Hosten
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
- INSERM UMR1144, Université Paris Cité, Paris, France
| | - Sébastien Goutal
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sarah Leterrier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Cassandre Corvo
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Louise Breuil
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
- INSERM UMR1144, Université Paris Cité, Paris, France
| | - Olivier Barret
- CEA, CNRS, Université Paris-Saclay, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-Aux-Roses, France
| | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Charles Truillet
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Nicolas Tournier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| |
Collapse
|
4
|
Roll W, Faust A, Hermann S, Schäfers M. Infection Imaging: Focus on New Tracers? J Nucl Med 2023; 64:59S-67S. [PMID: 37918846 DOI: 10.2967/jnumed.122.264869] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/31/2023] [Indexed: 11/04/2023] Open
Abstract
Infections account for relevant morbidity and mortality, especially if the cardiovascular system is affected. Clinical manifestations are often unspecific, resulting in a challenging diagnostic work-up. The use of molecular imaging methods, namely [18F]FDG PET and leukocyte scintigraphy, is increasingly recognized in recently published international guidelines. However, these 2 established methods focus on the host's immune response to the pathogen and are therefore virtually unable to differentiate infection from inflammation. Targeting the microorganism responsible for the infection directly with novel imaging agents is a promising strategy to overcome these limitations. In this review, we discuss clinically approved [18F]FDG PET with its advantages and limitations in cardiovascular infections, followed by new PET-based approaches for the detection of cardiovascular infections by bacteria-specific molecular imaging methods. A multitude of different targeting options has already been preclinically evaluated, but most still lack clinical translation. We give an overview not only on promising tracer candidates for noninvasive molecular imaging of infections but also on issues hampering clinical translation.
Collapse
Affiliation(s)
- Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany; and
| | - Andreas Faust
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany; and
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany; and
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| |
Collapse
|
5
|
Ordonez AA, Saupe F, Kasper CA, Turner ML, Parveen S, Flavahan K, Shin H, Artemov D, Ittig SJ, Jain SK. Imaging Tumor-Targeting Bacteria Using 18F-Fluorodeoxysorbitol Positron Emission Tomography. J Infect Dis 2023; 228:S291-S296. [PMID: 37788499 DOI: 10.1093/infdis/jiad077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Microbial-based cancer treatments are an emerging field, with multiple bacterial species evaluated in animal models and some advancing to clinical trials. Noninvasive bacteria-specific imaging approaches can potentially support the development and clinical translation of bacteria-based cancer treatments by assessing the tumor and off-target bacterial colonization. METHODS 18F-Fluorodeoxysorbitol (18F-FDS) positron emission tomography (PET), a bacteria-specific imaging approach, was used to visualize an attenuated strain of Yersinia enterocolitica, currently in clinical trials as a microbial-based cancer treatment, in murine models of breast cancer. RESULTS Y. enterocolitica demonstrated excellent 18F-FDS uptake in in vitro assays. Whole-body 18F-FDS PET demonstrated a significantly higher PET signal in tumors with Y. enterocolitica colonization compared to those not colonized, in murine models utilizing direct intratumor or intravenous administration of bacteria, which were confirmed using ex vivo gamma counting. Conversely, 18F-fluorodeoxyglucose (18F-FDG) PET signal was not different in Y. enterocolitica colonized versus uncolonized tumors. CONCLUSIONS Given that PET is widely used for the management of cancer patients, 18F-FDS PET could be utilized as a complementary approach supporting the development and clinical translation of Y. enterocolitica-based tumor-targeting bacterial therapeutics.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Falk Saupe
- T3 Pharmaceuticals AG, Allschwil, Switzerland
| | | | - Mitchell L Turner
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sadiya Parveen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelly Flavahan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hyunsoo Shin
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dmitri Artemov
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Chen X, Gallagher F, Sellmyer MA, Ordonez AA, Kjaer A, Ohliger M, Wilson DM, Jain SK. Visualizing Bacterial Infections With Novel Targeted Molecular Imaging Approaches. J Infect Dis 2023; 228:S249-S258. [PMID: 37788506 PMCID: PMC10547462 DOI: 10.1093/infdis/jiad078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
Although nearly a century has elapsed since the discovery of penicillin, bacterial infections remain a major global threat. Global antibiotic use resulted in an astounding 42 billion doses of antibiotics administered in 2015 with 128 billion annual doses expected by 2030. This overuse of antibiotics has led to the selection of multidrug-resistant "super-bugs," resulting in increasing numbers of patients being susceptible to life-threatening infections with few available therapeutic options. New clinical tools are therefore urgently needed to identify bacterial infections and monitor response to antibiotics, thereby limiting overuse of antibiotics and improving overall health. Next-generation molecular imaging affords unique opportunities to target and identify bacterial infections, enabling spatial characterization as well as noninvasive, temporal monitoring of the natural course of the disease and response to therapy. These emerging noninvasive imaging approaches could overcome several limitations of current tools in infectious disease, such as the need for biological samples for testing with their associated sampling bias. Imaging of living bacteria can also reveal basic biological insights about their behavior in vivo.
Collapse
Affiliation(s)
- Xueyi Chen
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ferdia Gallagher
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital–Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Ohliger
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Sorlin A, López-Álvarez M, Rabbitt SJ, Alanizi AA, Shuere R, Bobba KN, Blecha J, Sakhamuri S, Evans MJ, Bayles KW, Flavell RR, Rosenberg OS, Sriram R, Desmet T, Nidetzky B, Engel J, Ohliger MA, Fraser JS, Wilson DM. Chemoenzymatic Syntheses of Fluorine-18-Labeled Disaccharides from [ 18F] FDG Yield Potent Sensors of Living Bacteria In Vivo. J Am Chem Soc 2023; 145:17632-17642. [PMID: 37535945 PMCID: PMC10436271 DOI: 10.1021/jacs.3c03338] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 08/05/2023]
Abstract
Chemoenzymatic techniques have been applied extensively to pharmaceutical development, most effectively when routine synthetic methods fail. The regioselective and stereoselective construction of structurally complex glycans is an elegant application of this approach that is seldom applied to positron emission tomography (PET) tracers. We sought a method to dimerize 2-deoxy-[18F]-fluoro-d-glucose ([18F]FDG), the most common tracer used in clinical imaging, to form [18F]-labeled disaccharides for detecting microorganisms in vivo based on their bacteria-specific glycan incorporation. When [18F]FDG was reacted with β-d-glucose-1-phosphate in the presence of maltose phosphorylase, the α-1,4- and α-1,3-linked products 2-deoxy-[18F]-fluoro-maltose ([18F]FDM) and 2-deoxy-2-[18F]-fluoro-sakebiose ([18F]FSK) were obtained. This method was further extended with the use of trehalose (α,α-1,1), laminaribiose (β-1,3), and cellobiose (β-1,4) phosphorylases to synthesize 2-deoxy-2-[18F]fluoro-trehalose ([18F]FDT), 2-deoxy-2-[18F]fluoro-laminaribiose ([18F]FDL), and 2-deoxy-2-[18F]fluoro-cellobiose ([18F]FDC). We subsequently tested [18F]FDM and [18F]FSK in vitro, showing accumulation by several clinically relevant pathogens including Staphylococcus aureus and Acinetobacter baumannii, and demonstrated their specific uptake in vivo. Both [18F]FDM and [18F]FSK were stable in human serum with high accumulation in preclinical infection models. The synthetic ease and high sensitivity of [18F]FDM and [18F]FSK to S. aureus including methicillin-resistant (MRSA) strains strongly justify clinical translation of these tracers to infected patients. Furthermore, this work suggests that chemoenzymatic radiosyntheses of complex [18F]FDG-derived oligomers will afford a wide array of PET radiotracers for infectious and oncologic applications.
Collapse
Affiliation(s)
- Alexandre
M. Sorlin
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Marina López-Álvarez
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Sarah J. Rabbitt
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Aryn A. Alanizi
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Rebecca Shuere
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Kondapa Naidu Bobba
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Joseph Blecha
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Sasank Sakhamuri
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Michael J. Evans
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Kenneth W. Bayles
- Department
of Pathology and Microbiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Robert R. Flavell
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Oren S. Rosenberg
- Department
of Medicine University of California, San
Francisco, San Francisco, California 94158, United States
| | - Renuka Sriram
- Department
of Biotechnology, Ghent University, Gent B-9000, Belgium
| | - Tom Desmet
- Department
of Biotechnology, Ghent University, Gent B-9000, Belgium
| | - Bernd Nidetzky
- Institute
of Biotechnology and Biochemical Engineering, Graz University of Technology, Graz 8010, Austria
| | - Joanne Engel
- Department
of Biotechnology, Ghent University, Gent B-9000, Belgium
| | - Michael A. Ohliger
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
- Department
of Radiology Zuckerberg San Francisco General
Hospital, San Francisco, California 94110, United States
| | - James S. Fraser
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
| | - David M. Wilson
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
8
|
Sorlin AM, López-Álvarez M, Rabbitt SJ, Alanizi AA, Shuere R, Bobba KN, Blecha J, Sakhamuri S, Evans MJ, Bayles KW, Flavell RR, Rosenberg OS, Sriram R, Desmet T, Nidetzky B, Engel J, Ohliger MA, Fraser JS, Wilson DM. Chemoenzymatic syntheses of fluorine-18-labeled disaccharides from [ 18 F]FDG yield potent sensors of living bacteria in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.20.541529. [PMID: 37293043 PMCID: PMC10245702 DOI: 10.1101/2023.05.20.541529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chemoenzymatic techniques have been applied extensively to pharmaceutical development, most effectively when routine synthetic methods fail. The regioselective and stereoselective construction of structurally complex glycans is an elegant application of this approach, that is seldom applied to positron emission tomography (PET) tracers. We sought a method to dimerize 2-deoxy-[ 18 F]-fluoro-D-glucose ([ 18 F]FDG), the most common tracer used in clinical imaging, to form [ 18 F]-labeled disaccharides for detecting microorganisms in vivo based on their bacteria-specific glycan incorporation. When [ 18 F]FDG was reacted with β-D-glucose-1-phosphate in the presence of maltose phosphorylase, both the α-1,4 and α-1,3-linked products 2-deoxy-[ 18 F]-fluoro-maltose ([ 18 F]FDM) and 2-deoxy-2-[ 18 F]-fluoro-sakebiose ([ 18 F]FSK) were obtained. This method was further extended with the use of trehalose (α,α-1,1), laminaribiose (β-1,3), and cellobiose (β-1,4) phosphorylases to synthesize 2-deoxy-2-[ 18 F]fluoro-trehalose ([ 18 F]FDT), 2-deoxy-2-[ 18 F]fluoro-laminaribiose ([ 18 F]FDL), and 2-deoxy-2-[ 18 F]fluoro-cellobiose ([ 18 F]FDC). We subsequently tested [ 18 F]FDM and [ 18 F]FSK in vitro, showing accumulation by several clinically relevant pathogens including Staphylococcus aureus and Acinetobacter baumannii, and demonstrated their specific uptake in vivo. The lead sakebiose-derived tracer [ 18 F]FSK was stable in human serum and showed high uptake in preclinical models of myositis and vertebral discitis-osteomyelitis. Both the synthetic ease, and high sensitivity of [ 18 F]FSK to S. aureus including methicillin-resistant (MRSA) strains strongly justify clinical translation of this tracer to infected patients. Furthermore, this work suggests that chemoenzymatic radiosyntheses of complex [ 18 F]FDG-derived oligomers will afford a wide array of PET radiotracers for infectious and oncologic applications.
Collapse
|
9
|
Braams LM, Sijbesma JWA, Boersma HH, van Dijl JM, Elsinga PH, Glaudemans AWJM, Slart RHJA, van Oosten M. Preclinical evaluation of 2-[ 18F]fluorodeoxysorbitol as a tracer for targeted imaging of Enterobacterales infection. Int J Med Microbiol 2023; 313:151581. [PMID: 37209590 DOI: 10.1016/j.ijmm.2023.151581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023] Open
Abstract
Fluorine-18-fluorodeoxyglucose ([18F]FDG) positron emission tomography (18F-FDG-PET) is widely used for the detection of inflammatory and infectious diseases. Although this modality has proven to be a useful diagnostic tool, reliable distinction of bacterial infection from sterile inflammation or even from a malignancy remains challenging. Therefore, there is a need for bacteria-specific tracers for PET imaging that facilitate a reliable distinction of bacterial infection from other pathology. The present study was aimed at exploring the potential of 2-[18F]-fluorodeoxysorbitol ([18F]FDS) as a tracer for detection of Enterobacterales infections. Sorbitol is a sugar alcohol that is commonly metabolized by bacteria of the Enterobacterales order, but not by mammalian cells, which makes it an attractive candidate for targeted bacterial imaging. The latter is important in view of the serious clinical implications of infections caused by Enterobacterales. Here we demonstrate that sorbitol-based PET can be applied to detect a broad range of clinical bacterial isolates not only in vitro, but also in blood and ascites samples from patients suffering from Enterobacterales infections. Notably, the possible application of [18F]FDS is not limited to Enterobacterales since Pseudomonas aeruginosa and Corynebacterium jeikeium also showed substantial uptake of this tracer. We conclude that [18F]FDS is a promising tracer for PET-imaging of infections caused by a group of bacteria that can cause serious invasive disease.
Collapse
Affiliation(s)
- Lisanne M Braams
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands
| | - Jürgen W A Sijbesma
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands
| | - Hendrikus H Boersma
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands
| | - Jan Maarten van Dijl
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands.
| | - Philip H Elsinga
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands
| | - Andor W J M Glaudemans
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands
| | - Riemer H J A Slart
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands; TechMed Centre, Department of Biomedical Photonic Imaging, University of Twente, PO box 217, 7500 AE, Enschede, the Netherlands
| | - Marleen van Oosten
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Hanzeplein 1 PO box 30001, 9700RB Groningen, the Netherlands
| |
Collapse
|
10
|
Abstract
Bacterial infections are a major threat to human health. Rapid and accurate diagnosis of bacterial infections is essential for early interventions and rational use of antibiotic treatments. However, antibiotics are often initiated empirically while diagnostic tests are being performed. Moreover, traditional diagnostic tools, namely microscopy, microbiology and molecular techniques, are dependent upon sampling suspected sites of infection, and then performing tests. This approach is often invasive, labor intensive, time consuming, and subject to the uncertainties of incorrect sampling and contamination. There are currently no imaging approaches for the specific detection of bacterial infections. Therefore, there is a need for new noninvasive approaches to detect, localize and monitor bacterial infections with high sensitivity and specificity.
Collapse
Affiliation(s)
- Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
11
|
Rua M, Simón JA, Collantes M, Ecay M, Leiva J, Carmona-Torre F, Ramos R, Pareja F, Pulagam KR, Llop J, Del Pozo JL, Peñuelas I. Infection-specific PET imaging with 18F-fluorodeoxysorbitol and 2-[ 18F]F-ρ-aminobenzoic acid: An extended diagnostic tool for bacterial and fungal diseases. Front Microbiol 2023; 14:1094929. [PMID: 36760503 PMCID: PMC9905739 DOI: 10.3389/fmicb.2023.1094929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Suspected infectious diseases located in difficult-to-access sites can be challenging due to the need for invasive procedures to isolate the etiological agent. Positron emission tomography (PET) is a non-invasive imaging technology that can help locate the infection site. The most widely used radiotracer for PET imaging (2-deoxy-2[18F] fluoro-D-glucose: [18F]FDG) shows uptake in both infected and sterile inflammation. Therefore, there is a need to develop new radiotracers able to specifically detect microorganisms. Methods We tested two specific radiotracers: 2-deoxy-2-[18F]-fluoro-D-sorbitol ([18F]FDS) and 2-[18F]F-ρ-aminobenzoic acid ([18F]FPABA), and also developed a simplified alternative of the latter for automated synthesis. Clinical and reference isolates of bacterial and yeast species (19 different strains in all) were tested in vitro and in an experimental mouse model of myositis infection. Results and discussion Non-lactose fermenters (Pseudomonas aeruginosa and Stenotrophomonas maltophilia) were unable to take up [18F]FDG in vitro. [18F]FDS PET was able to visualize Enterobacterales myositis infection (i.e., Escherichia coli) and to differentiate between yeasts with differential assimilation of sorbitol (i.e., Candida albicans vs. Candida glabrata). All bacteria and yeasts tested were detected in vitro by [18F]FPABA. Furthermore, [18F]FPABA was able to distinguish between inflammation and infection in the myositis mouse model (E. coli and Staphylococcus aureus) and could be used as a probe for a wide variety of bacterial and fungal species.
Collapse
Affiliation(s)
- Marta Rua
- Clinical Microbiology Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Jon Ander Simón
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - María Collantes
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain,*Correspondence: María Collantes, ✉
| | - Margarita Ecay
- Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - José Leiva
- Clinical Microbiology Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Francisco Carmona-Torre
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,Infectious Diseases Division, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rocío Ramos
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - Félix Pareja
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - Krishna R. Pulagam
- Basque Research and Technology Alliance (BRTA), CIC BiomaGUNE, San Sebastián, Spain
| | - Jordi Llop
- Basque Research and Technology Alliance (BRTA), CIC BiomaGUNE, San Sebastián, Spain
| | - José Luis Del Pozo
- Clinical Microbiology Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,Infectious Diseases Division, Clínica Universidad de Navarra, Pamplona, Spain
| | - Iván Peñuelas
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain,Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
12
|
Campisciano G, Biffi S. Microbiota in vivo imaging approaches to study host-microbe interactions in preclinical and clinical setting. Heliyon 2022; 8:e12511. [PMID: 36593827 PMCID: PMC9803719 DOI: 10.1016/j.heliyon.2022.e12511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/14/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
In vivo imaging in preclinical and clinical settings can enhance knowledge of the host-microbiome interactions. Imaging techniques are a crucial node between findings at the molecular level and clinical implementation in diagnostics and therapeutics. The purpose of this study was to review existing knowledge on the microbiota in the field of in vivo imaging and provide guidance for future research, emphasizing the critical role that molecular imaging plays in increasing understanding of the host-microbe interaction. Preclinical microbiota animal models lay the foundation for the clinical translatability of novel microbiota-based therapeutics. Adopting animal models in which factors such as host genetic landscape, microbiota profile, and diet can be controlled enables investigating how the microbiota contributes to immunological dysregulation and inflammatory disorders. Current preclinical imaging of gut microbiota relies on models where the bacteria can be isolated, labelled, and re-administered. In vivo, optical imaging, ultrasound and magnetic resonance imaging define the bacteria's biodistribution in preclinical models, whereas nuclear imaging investigates bacterial metabolic activity. For the clinical investigation of microbe-host interactions, molecular nuclear imaging is increasingly becoming a promising approach. Future microbiota research should develop selective imaging probes to investigate in vivo microbiota profiles and individual strains of specific microbes. Preclinical knowledge can be translated into the molecular imaging field with great opportunities for studying the microbiome.
Collapse
Affiliation(s)
- Giuseppina Campisciano
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo Via dell'Istria 65/1, 34137, Trieste, Italy
| | - Stefania Biffi
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo Via dell'Istria 65/1, 34137, Trieste, Italy
| |
Collapse
|
13
|
Molecular Imaging of Ultrasound-Mediated Blood-Brain Barrier Disruption in a Mouse Orthotopic Glioblastoma Model. Pharmaceutics 2022; 14:pharmaceutics14102227. [PMID: 36297663 PMCID: PMC9610067 DOI: 10.3390/pharmaceutics14102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and malignant primary brain tumor. The blood-brain barrier (BBB) limits the therapeutic options available to tackle this incurable tumor. Transient disruption of the BBB by focused ultrasound (FUS) is a promising and safe approach to increase the brain and tumor concentration of drugs administered systemically. Non-invasive, sensitive, and reliable imaging approaches are required to better understand the impact of FUS on the BBB and brain microenvironment. In this study, nuclear imaging (SPECT/CT and PET/CT) was used to quantify neuroinflammation 48 h post-FUS and estimate the influence of FUS on BBB opening and tumor growth in vivo. BBB disruptions were performed on healthy and GBM-bearing mice (U-87 MG xenograft orthotopic model). The BBB recovery kinetics were followed and quantified by [99mTc]Tc-DTPA SPECT/CT imaging at 0.5 h, 3 h and 24 h post-FUS. The absence of neuroinflammation was confirmed by [18F]FDG PET/CT imaging 48 h post-FUS. The presence of the tumor and its growth were evaluated by [68Ga]Ga-RGD2 PET/CT imaging and post-mortem histological analysis, showing that tumor growth was not influenced by FUS. In conclusion, molecular imaging can be used to evaluate the time frame for systemic treatment combined with transient BBB opening and to test its efficacy over time.
Collapse
|
14
|
In vivo imaging of invasive aspergillosis with 18F-fluorodeoxysorbitol positron emission tomography. Nat Commun 2022; 13:1926. [PMID: 35395822 PMCID: PMC8993802 DOI: 10.1038/s41467-022-29553-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 03/23/2022] [Indexed: 11/08/2022] Open
Abstract
Invasive aspergillosis is a critical complication in immunocompromised patients with hematologic malignancies or with viral pneumonia caused by influenza virus or SARS‑CoV‑2. Although early and accurate diagnosis of invasive aspergillosis can maximize clinical outcomes, current diagnostic methods are time-consuming and poorly sensitive. Here, we assess the ability of 2-deoxy-2-18F-fluorosorbitol (18F-FDS) positron emission tomography (PET) to specifically and noninvasively detect Aspergillus infections. We show that 18F-FDS PET can be used to visualize Aspergillus fumigatus infection of the lungs, brain, and muscles in mouse models. In particular, 18F-FDS can distinguish pulmonary aspergillosis from Staphylococcus aureus infection, both of which induce pulmonary infiltrates in immunocompromised patients. Thus, our results indicate that the combination of 18F-FDS PET and appropriate clinical information may be useful in the differential diagnosis and localization of invasive aspergillosis.
Collapse
|
15
|
Evaluation of 2-[ 18F]-Fluorodeoxysorbitol PET Imaging in Preclinical Models of Aspergillus Infection. J Fungi (Basel) 2021; 8:jof8010025. [PMID: 35049965 PMCID: PMC8780649 DOI: 10.3390/jof8010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
Despite increasing associated mortality and morbidity, the diagnosis of fungal infections, especially with Aspergillus fumigatus (A. fumigatus), remains challenging. Based on known ability of Aspergillus species to utilize sorbitol, we evaluated 2-[18F]-fluorodeoxysorbitol (FDS), a recently described Enterobacterales imaging ligand, in animal models of A. fumigatus infection, in comparison with 2-[18F]-fluorodeoxyglucose (FDG). In vitro assays showed slightly higher 3H-sorbitol uptake by live compared with heat-killed A. fumigatus. However, this was 10.6-fold lower than E. coli uptake. FDS positron emission tomography (PET) imaging of A. fumigatus pneumonia showed low uptake in infected lungs compared with FDG (0.290 ± 0.030 vs. 8.416 ± 0.964 %ID/mL). This uptake was higher than controls (0.098 ± 0.008 %ID/mL) and minimally higher than lung inflammation (0.167 ± 0.007 %ID/mL). In the myositis models, FDS uptake was highest in live E. coli infections. Uptake was low in A. fumigatus myositis model and only slightly higher in live compared with the heat-killed side. In conclusion, we found low uptake of 3H-sorbitol and FDS by A. fumigatus cultures and infection models compared with E. coli, likely due to the need for induction of sorbitol dehydrogenase by sorbitol. Our findings do not support FDS as an Aspergillus imaging agent. At this point, FDS remains more selective for imaging Gram-negative Enterobacterales.
Collapse
|
16
|
More S, Marakalala MJ, Sathekge M. Tuberculosis: Role of Nuclear Medicine and Molecular Imaging With Potential Impact of Neutrophil-Specific Tracers. Front Med (Lausanne) 2021; 8:758636. [PMID: 34957144 PMCID: PMC8703031 DOI: 10.3389/fmed.2021.758636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/03/2021] [Indexed: 01/02/2023] Open
Abstract
With Tuberculosis (TB) affecting millions of people worldwide, novel imaging modalities and tools, particularly nuclear medicine and molecular imaging, have grown with greater interest to assess the biology of the tuberculous granuloma and evolution thereof. Much early work has been performed at the pre-clinical level using gamma single photon emission computed tomography (SPECT) agents exploiting certain characteristics of Mycobacterium tuberculosis (MTb). Both antituberculous SPECT and positron emission tomography (PET) agents have been utilised to characterise MTb. Other PET tracers have been utilised to help to characterise the biology of MTb (including Gallium-68-labelled radiopharmaceuticals). Of all the tracers, 2-[18F]FDG has been studied extensively over the last two decades in many aspects of the treatment paradigm of TB: at diagnosis, staging, response assessment, restaging, and in potentially predicting the outcome of patients with latent TB infection. Its lower specificity in being able to distinguish different inflammatory cell types in the granuloma has garnered interest in reviewing more specific agents that can portend prognostic implications in the management of MTb. With the neutrophil being a cell type that portends this poorer prognosis, imaging this cell type may be able to answer more accurately questions relating to the tuberculous granuloma transmissivity and may help in characterising patients who may be at risk of developing active TB. The formyl peptide receptor 1(FPR1) expressed by neutrophils is a key marker in this process and is a potential target to characterise these areas. The pre-clinical work regarding the role of radiolabelled N-cinnamoyl –F-(D) L – F – (D) –L F (cFLFLF) (which is an antagonist for FPR1) using Technetium 99m-labelled conjugates and more recently radiolabelled with Gallium-68 and Copper 64 is discussed. It is the hope that further work with this tracer may accelerate its potential to be utilised in responding to many of the current diagnostic dilemmas and challenges in TB management, thereby making the tracer a translatable option in routine clinical care.
Collapse
Affiliation(s)
- Stuart More
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
- *Correspondence: Stuart More
| | - Mohlopheni J. Marakalala
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Michael Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
| |
Collapse
|
17
|
Mota F, De Jesus P, Jain SK. Kit-based synthesis of 2-deoxy-2-[ 18F]-fluoro-D-sorbitol for bacterial imaging. Nat Protoc 2021; 16:5274-5286. [PMID: 34686858 PMCID: PMC8611807 DOI: 10.1038/s41596-021-00613-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023]
Abstract
Clinically available imaging tools for diagnosing infections rely on structural changes in the affected tissues. They therefore lack specificity and cannot differentiate between oncologic, inflammatory and infectious processes. We have developed 2-deoxy-2-[18F]fluoro-D-sorbitol (18F-FDS) as an imaging agent to visualize infections caused by Enterobacterales, which represent the largest group of bacterial pathogens in humans and are responsible for severe infections, often resulting in sepsis or death. A clinical study in 26 prospectively enrolled patients demonstrated that 18F-FDS positron emission tomography (PET) was safe, and could detect and localize infections due to drug-susceptible or multi-drug-resistant Enterobacterales strains as well as differentiate them from other pathologies (sterile inflammation or cancer). 18F-FDS is cleared almost exclusively through renal filtration and has also shown potential as a PET agent for functional renal imaging. Since most PET radionuclides have a short half-life, maximal clinical impact will require fast, on-demand synthesis with limited infrastructure and personnel. To meet this demand, we developed a kit-based solid phase method that uses commercially and widely available 2-deoxy-2-[18F]fluoro-D-glucose as the precursor and allows 18F-FDS to be produced and purified in one step at room temperature. The 18F-FDS kit consists of a solid-phase extraction cartridge packed with solid supported borohydride (MP-borohydride), which can be attached to a second cartridge to reduce pH. We evaluated the effects of different solid supported borohydride reagents, cartridge size, starting radioactivity, volumes and flow rates in the radiochemical yield and purity. The optimized protocol can be completed in <30 min and allows the synthesis of 18F-FDS in >70% radiochemical yield and >90% radiochemical purity.
Collapse
Affiliation(s)
- Filipa Mota
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patricia De Jesus
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Abstract
Most studies of gut microbiota have focused on relationships between a specific disease and the presence/abundance of one or a few bacterial species/genera. Whether the spatial and temporal distribution of gut microbiota, as a whole, affects or correlates with health is unknown, largely due to the absence of tools for dynamically monitoring the overall gut microbiota landscape inside living subjects. Here, we describe a novel, noninvasive, live imaging method for gut microbiota using 2-deoxy-2-[18F]fluoro-d-sorbitol (18F-FDS), a compound that specifically labeled gut bacteria in mice and hamsters following oral administration. Positron emission tomography-computed tomography (PET-CT) scanning showed that the radiolabel signal was concentrated in the gut (especially the large intestine), was absent when mice gut microbiota was depleted by antibiotic treatment, and was restored after transplanting antibiotic-treated mice with a fecal or probiotic bacterial mixture. Thus, 18F-FDS images microbiota, not gut tissue. The tissue distribution of 18F-FDS was the highest in the gut (∼3-fold higher than average), in contrast to 2-deoxy-2-[18F]fluoro-d-glucose, which concentrated in brain and many other organs. 2-[18F]fluoro-aminobenzoic acid, another bacterium-specific radioactive tracer, was unsuited for gut microbiota imaging due to unexpected stomach retention following oral administration. When similar gut microbiota imaging was done with hamsters, the spatial resolution increased significantly over that with mice, suggesting that even higher spatial resolution can be achieved with humans or large animals. Thus, our work establishes a new tool for noninvasive, live imaging of gut microbiota; the new tool may enable exploration of relationships between gut microbiota landscape and diseases in clinical settings. IMPORTANCE Gut microbiota dysbiosis correlates with many diseases, but such correlations derive mostly from relationships between one or a few bacteria and a particular disease. Since microbiota resemble complex forest ecosystems more closely than individual patches of trees, the overall landscape (spatial and temporal distribution) of gut bacteria may also affect/reflect disease development. Such a possibility has not been explored due to a lack of tools for directly visualizing natural landscape patterns of gut microbiota. The present work identified 2-deoxy-2-[18F]fluoro-d-sorbitol as a gut microbiota-specific radioactive tracer and developed a novel PET-CT scan-based imaging method that enables noninvasive, real-time imaging of the overall gut bacterial landscape. The method showed increased spatial resolution when hamsters replaced mice, suggesting that even higher spatial resolution could be achieved with larger animals such as humans. This novel technology establishes the feasibility of investigating spatial-temporal distribution dynamics of gut microbiota with many human diseases.
Collapse
|
19
|
Hugon G, Goutal S, Dauba A, Breuil L, Larrat B, Winkeler A, Novell A, Tournier N. [ 18F]2-Fluoro-2-deoxy-sorbitol PET Imaging for Quantitative Monitoring of Enhanced Blood-Brain Barrier Permeability Induced by Focused Ultrasound. Pharmaceutics 2021; 13:pharmaceutics13111752. [PMID: 34834167 PMCID: PMC8621256 DOI: 10.3390/pharmaceutics13111752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 01/18/2023] Open
Abstract
Focused ultrasound in combination with microbubbles (FUS) provides an effective means to locally enhance the delivery of therapeutics to the brain. Translational and quantitative imaging techniques are needed to noninvasively monitor and optimize the impact of FUS on blood-brain barrier (BBB) permeability in vivo. Positron-emission tomography (PET) imaging using [18F]2-fluoro-2-deoxy-sorbitol ([18F]FDS) was evaluated as a small-molecule (paracellular) marker of blood-brain barrier (BBB) integrity. [18F]FDS was straightforwardly produced from chemical reduction of commercial [18F]2-deoxy-2-fluoro-D-glucose. [18F]FDS and the invasive BBB integrity marker Evan’s blue (EB) were i.v. injected in mice after an optimized FUS protocol designed to generate controlled hemispheric BBB disruption. Quantitative determination of the impact of FUS on the BBB permeability was determined using kinetic modeling. A 2.2 ± 0.5-fold higher PET signal (n = 5; p < 0.01) was obtained in the sonicated hemisphere and colocalized with EB staining observed post mortem. FUS significantly increased the blood-to-brain distribution of [18F]FDS by 2.4 ± 0.8-fold (VT; p < 0.01). Low variability (=10.1%) of VT values in the sonicated hemisphere suggests reproducibility of the estimation of BBB permeability and FUS method. [18F]FDS PET provides a readily available, sensitive and reproducible marker of BBB permeability to noninvasively monitor the extent of BBB disruption induced by FUS in vivo.
Collapse
Affiliation(s)
- Gaëlle Hugon
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Sébastien Goutal
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Ambre Dauba
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Louise Breuil
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Benoit Larrat
- CNRS, CEA, DRF/JOLIOT/NEUROSPIN/BAOBAB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France;
| | - Alexandra Winkeler
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Anthony Novell
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Nicolas Tournier
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
- Correspondence:
| |
Collapse
|
20
|
Ordonez AA, Wintaco LM, Mota F, Restrepo AF, Ruiz-Bedoya CA, Reyes CF, Uribe LG, Abhishek S, D'Alessio FR, Holt DP, Dannals RF, Rowe SP, Castillo VR, Pomper MG, Granados U, Jain SK. Imaging Enterobacterales infections in patients using pathogen-specific positron emission tomography. Sci Transl Med 2021; 13:13/589/eabe9805. [PMID: 33853931 DOI: 10.1126/scitranslmed.abe9805] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/10/2021] [Indexed: 12/29/2022]
Abstract
Enterobacterales represent the largest group of bacterial pathogens in humans and are responsible for severe, deep-seated infections, often resulting in sepsis or death. They are also a prominent cause of multidrug-resistant (MDR) infections, and some species are recognized as biothreat pathogens. Tools for noninvasive, whole-body analysis that can localize a pathogen with specificity are needed, but no such technology currently exists. We previously demonstrated that positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-d-sorbitol (18F-FDS) can selectively detect Enterobacterales infections in murine models. Here, we demonstrate that uptake of 18F-FDS by bacteria occurs via a metabolically conserved sorbitol-specific pathway with rapid in vitro 18F-FDS uptake noted in clinical strains, including MDR isolates. Whole-body 18F-FDS PET/computerized tomography (CT) in 26 prospectively enrolled patients with either microbiologically confirmed Enterobacterales infection or other pathologies demonstrated that 18F-FDS PET/CT was safe, could rapidly detect and localize Enterobacterales infections due to drug-susceptible or MDR strains, and differentiated them from sterile inflammation or cancerous lesions. Repeat imaging in the same patients monitored antibiotic efficacy with decreases in PET signal correlating with clinical improvement. To facilitate the use of 18F-FDS, we developed a self-contained, solid-phase cartridge to rapidly (<10 min) formulate ready-to-use 18F-FDS from commercially available 2-deoxy-2-[18F]fluoro-d-glucose (18F-FDG) at room temperature. In a hamster model, 18F-FDS PET/CT also differentiated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia from secondary Klebsiella pneumoniae pneumonia-a leading cause of complications in hospitalized patients with COVID-19. These data support 18F-FDS as an innovative and readily available, pathogen-specific PET technology with clinical applications.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Luz M Wintaco
- Department of Nuclear Medicine, Hospital Internacional de Colombia, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia.,Biomedical and Biological Sciences Graduate Program, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| | - Filipa Mota
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andres F Restrepo
- Department of Internal Medicine, Hospital Internacional de Colombia, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia
| | - Camilo A Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Carlos F Reyes
- Department of Critical Care, Hospital Internacional de Colombia, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia
| | - Luis G Uribe
- Department of Infectious Diseases, Hospital Internacional de Colombia, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia
| | - Sudhanshu Abhishek
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Franco R D'Alessio
- Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel P Holt
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert F Dannals
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Victor R Castillo
- Bioengineering Research Group, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ulises Granados
- Department of Nuclear Medicine, Hospital Internacional de Colombia, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia. .,Biomedical and Translational Research Group, Fundación Cardiovascular de Colombia, Piedecuesta 681017, Colombia
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. .,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
21
|
Jødal L, Afzelius P, Alstrup AKO, Jensen SB. Radiotracers for Bone Marrow Infection Imaging. Molecules 2021; 26:3159. [PMID: 34070537 PMCID: PMC8198735 DOI: 10.3390/molecules26113159] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Radiotracers are widely used in medical imaging, using techniques of gamma-camera imaging (scintigraphy and SPECT) or positron emission tomography (PET). In bone marrow infection, there is no single routine test available that can detect infection with sufficiently high diagnostic accuracy. Here, we review radiotracers used for imaging of bone marrow infection, also known as osteomyelitis, with a focus on why these molecules are relevant for the task, based on their physiological uptake mechanisms. The review comprises [67Ga]Ga-citrate, radiolabelled leukocytes, radiolabelled nanocolloids (bone marrow) and radiolabelled phosphonates (bone structure), and [18F]FDG as established radiotracers for bone marrow infection imaging. Tracers that are under development or testing for this purpose include [68Ga]Ga-citrate, [18F]FDG, [18F]FDS and other non-glucose sugar analogues, [15O]water, [11C]methionine, [11C]donepezil, [99mTc]Tc-IL-8, [68Ga]Ga-Siglec-9, phage-display selected peptides, and the antimicrobial peptide [99mTc]Tc-UBI29-41 or [68Ga]Ga-NOTA-UBI29-41. CONCLUSION Molecular radiotracers allow studies of physiological processes such as infection. None of the reviewed molecules are ideal for the imaging of infections, whether bone marrow or otherwise, but each can give information about a separate aspect such as physiology or biochemistry. Knowledge of uptake mechanisms, pitfalls, and challenges is useful in both the use and development of medically relevant radioactive tracers.
Collapse
Affiliation(s)
- Lars Jødal
- Department of Nuclear Medicine, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
| | - Pia Afzelius
- Zealand Hospital, Køge, Copenhagen University Hospital, DK-4600 Køge, Denmark;
| | - Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine & PET, Aarhus University Hospital, DK-8200 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Svend Borup Jensen
- Department of Nuclear Medicine, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
- Department of Chemistry and Biosciences, Aalborg University, DK-9220 Aalborg, Denmark
| |
Collapse
|
22
|
Hasegawa K, Koshino K, Higuchi T. Facile synthesis of 2-deoxy-2-[ 18 F]fluorosorbitol using sodium borohydride on aluminum oxide. J Labelled Comp Radiopharm 2021; 64:40-46. [PMID: 33063893 DOI: 10.1002/jlcr.3887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 11/09/2022]
Abstract
2-Deoxy-2-[18 F]fluorosorbitol (18 F-FDS) has become increasingly useful in functional renal imaging. FDS is synthesized by the one-step reduction of 2-deoxy-2-[18 F]fluoroglucose (18 F-FDG). To develop a more simple and rapid procedure for 18 F-FDS synthesis, we examined reduction reactions with solid-supported NaBH4 . Synthetic yields using BH4 -IRA400 (polymer-based matrix) and NaBH4 -Al2 O3 (clay-based matrix) as solid-supported reagents were compared. NaBH4 -Al2 O3 was found to be far superior to BH4 -IRA400 in the FDG reduction reaction. IRA 400 was not suitable for this reaction because it adsorbs FDG, in addition to glucose, with no FDS synthesized when using BH4 -IRA400. By contrast, NaBH4 -Al2 O3 only required a filtration as workup, affording FDS in 90% yield after a total of 10 min. NaBH4 on alumina was readily consumed in the reaction within 1 min, regardless of the amount used, by simply stirring with a vortex mixer. Complicated procedures, such as microwave irradiation, were not necessary. This simple operation will allow kit formulation and is suitable for radiosynthesis. In conclusion, clay-supported reagents showed low absorption and were time saving, which are highly compatible with 18 F-FDS synthesis.
Collapse
Affiliation(s)
- Koki Hasegawa
- Center for Instrumental Analysis, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kazuhiro Koshino
- Department of Systems and Informatics, Hokkaido Information University, Ebetsu, Japan
| | - Takahiro Higuchi
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Giron MC, Mazzi U. Molecular imaging of microbiota-gut-brain axis: searching for the right targeted probe for the right target and disease. Nucl Med Biol 2021; 92:72-77. [PMID: 33262001 DOI: 10.1016/j.nucmedbio.2020.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022]
Abstract
The highly bidirectional dialogue between the gut and the brain is markedly stimulated and influenced by the microbiome through integrated neuroendocrine, neurological and immunological processes. Gut microbiota itself communicate with the host producing hormonal intermediates, metabolites, proteins, and toxins responsible for a variety of biochemical and functional inputs, thereby shaping host homeostasis. Indeed, a dysregulated microbiota-gut-brain axis might be the origin of many neuroimmune-mediated disorders, e.g. autism, multiple sclerosis, depression, Alzheimer's and Parkinson's disease, which appear months or even years prior to a diagnosis, corroborating the theory that the pathological process is spread from the gut to the brain. A much deeper comprehension of how commensal microbe can be manipulated to interfere with disease progression is crucial for developing new strategies to diagnose and treat diseases. In recent years, the potential of positron-emission-tomography (PET) in the field of bacteria detection has gained attention. The uptake of several PET tracers has been evaluated to investigate infection pathophysiology, e.g. sterile or pathogen-mediated infection, monitoring of progression, or as a surrogate endpoint in clinical trials. In this minireview, we briefly describe the role of microbiome-gut-brain axis in health and disease and we discuss the imaging modalities and agents that could be applied to study the dynamic interactions between microbiome, gut and brain. These are key aspects in understanding the biochemical lexicon underpinning the microbiome-host crosstalk that would enable the development of diagnostics and therapeutics by targeting the human microbiota.
Collapse
Affiliation(s)
- Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy.
| | - Ulderico Mazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| |
Collapse
|
24
|
Ordoñez AA, Jain SK. Imaging of Bacterial Infections. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00089-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
25
|
Polvoy I, Flavell RR, Rosenberg OS, Ohliger MA, Wilson DM. Nuclear Imaging of Bacterial Infection: The State of the Art and Future Directions. J Nucl Med 2020; 61:1708-1716. [PMID: 32764120 DOI: 10.2967/jnumed.120.244939] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Increased mortality rates from infectious diseases is a growing public health concern. Successful management of acute bacterial infections requires early diagnosis and treatment, which are not always easy to achieve. Structural imaging techniques such as CT and MRI are often applied to this problem. However, these methods generally rely on secondary inflammatory changes and are frequently not specific to infection. The use of nuclear medicine techniques can add crucial complementary information, allowing visualization of infectious pathophysiology beyond morphologic imaging. This review will discuss the current structural and functional imaging techniques used for the diagnosis of bacterial infection and their roles in different clinical scenarios. We will also present several new radiotracers in development, with an emphasis on probes targeting bacteria-specific metabolism. As highlighted by the current coronavirus disease 2019 epidemic, caused by the novel severe acute respiratory syndrome coronavirus 2, similar thinking may apply in imaging viral pathogens; for this case, prominent effects on host proteins, most notably angiotensin-converting enzyme 2, might also provide worthwhile imaging targets.
Collapse
Affiliation(s)
- Ilona Polvoy
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Oren S Rosenberg
- Department of Medicine, University of California, San Francisco, San Francisco, California; and
| | - Michael A Ohliger
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,Department of Radiology, Zuckerberg San Francisco General Hospital, San Francisco, California
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| |
Collapse
|
26
|
Kong Z, Wang Y, Ma W, Cheng X. Role of 18F-fluorodeoxyglucose (FDG) and 18F-2-fluorodeoxy sorbitol (FDS) in autoimmune hypophysitis: a case report. BMC Endocr Disord 2020; 20:84. [PMID: 32517690 PMCID: PMC7285776 DOI: 10.1186/s12902-020-00567-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/03/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Autoimmune hypophysitis is a rare disease characterized by the infiltration of lymphocytic cells into the pituitary gland. 18F-fluorodeoxyglucose (FDG) and 18F-2-fluorodeoxy sorbitol (FDS) positron emission tomography (PET) are well-established and emerging techniques, respectively, which may aid in the diagnosis and classification of autoimmune hypophysitis. CASE PRESENTATION Here, we report a 40-year-old female diagnosed with central diabetes insipidus and multiple pituitary hormone deficiencies, and MRI revealed homogeneous signals in the pituitary gland as well as thickened in the pituitary stalk. FDG PET localized the pituitary and pituitary stalk lesions and displayed an SUVmax of 5.5. FDS, a sensitive radiotracer for bacterial infections but remains unproven under aseptic inflammation, also demonstrated elevated radioactivity, with an SUVmax of 1.1 at 30 min and 0.73 at 120 min. Transnasal biopsy suggested a diagnosis of autoimmune hypophysitis, and the patient displayed radiological and clinical improvement after treatment with glucocorticoids and hormone replacement. CONCLUSIONS Autoimmune hypophysitis can display elevated FDG uptake, which aids in the localization of the lesions. In addition to revealing bacterial infection specifically, FDS can also accumulate under autoimmune conditions, suggesting that it could serve as a potential radiotracer for both bacterial and aseptic inflammation. TRIAL REGISTRATION The patient was enrolled in study NCT02450942 (clinicaltrials.gov, Registered May 21, 2015).
Collapse
Affiliation(s)
- Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Xin Cheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| |
Collapse
|
27
|
Kang SR, Jo EJ, Nguyen VH, Zhang Y, Yoon HS, Pyo A, Kim DY, Hong Y, Bom HS, Min JJ. Imaging of tumor colonization by Escherichia coli using 18F-FDS PET. Am J Cancer Res 2020; 10:4958-4966. [PMID: 32308761 PMCID: PMC7163454 DOI: 10.7150/thno.42121] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/16/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor-targeting bacteria have been actively investigated as a new therapeutic tool for solid tumors. However, in vivo imaging of tumor-targeting bacteria has not been fully established. 18F-fluorodeoxysorbitol (FDS) positron emission tomography (PET) is known to be capable of imaging Gram-negative Enterobacteriaceae infection. In the present study, we aimed to validate the use of 18F-FDS PET for visualization of the colonization and proliferation of tumor-targeting Escherichia coli (E. coli) MG1655 in mouse tumor models. Methods: E. coli (5 × 107 colony forming unit) were injected intravenously into BALB/c mice bearing mouse colon cancer (CT26). Before and 1, 3, and 5 days after the bacterial injection, PET imaging was performed following i.v. injection of approximately 7.4 MBq of 18F-FDS. Regions of interest were drawn in the engrafted tumor and normal organs including the heart, liver, lung, brain, muscle, and intestine. Semiquantitative analysis was performed using maximum standardized uptake value (SUVmax). Results: 18F-FDS uptake was significantly higher in tumors colonized by live E. coli MG1655 than in uncolonized tumors (p < 0.001). The PET signals in the colonized tumors at 3 days after bacterial injection were 3.1-fold higher than those in the uncolonized tumors. Tumoral 18F-FDS uptake correlated very strongly with the number of E. coli in tumors (r = 0.823, p < 0.0001). Cross sectional analysis of autoradiography, bioluminescence, and pathology revealed that the 18F-FDS uptake sites in tumors matched the locations of E. coli MG1655. Conclusion: In conclusion, 18F-FDS PET is expected to be useful for the semiquantitative visualization of tumor-targeting bacteria when bacterial cancer therapy is performed using Gram-negative Enterobacteriaceae such as E. coli.
Collapse
|
28
|
Mota F, Ordonez AA, Firth G, Ruiz-Bedoya CA, Ma MT, Jain SK. Radiotracer Development for Bacterial Imaging. J Med Chem 2020; 63:1964-1977. [PMID: 32048838 DOI: 10.1021/acs.jmedchem.9b01623] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bacterial infections remain a major threat to humanity and are a leading cause of death and disability. Antimicrobial resistance has been declared as one of the top ten threats to human health by the World Health Organization, and new technologies are urgently needed for the early diagnosis and monitoring of deep-seated and complicated infections in hospitalized patients. This review summarizes the radiotracers as applied to imaging of bacterial infections. We summarize the recent progress in the development of pathogen-specific imaging and the application of radiotracers in understanding drug pharmacokinetics as well as the local biology at the infection sites. We also highlight the opportunities for medicinal chemists in radiotracer development for bacterial infections, with an emphasis on target selection and radiosynthetic approaches. Imaging of infections is an emerging field. Beyond clinical applications, these technologies could provide unique insights into disease pathogenesis and expedite bench-to-bedside translation of new therapeutics.
Collapse
Affiliation(s)
- Filipa Mota
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - George Firth
- School of Biomedical Engineering and Imaging Sciences, St. Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Camilo A Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, St. Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
29
|
Russelli L, Martinelli J, De Rose F, Reder S, Herz M, Schwaiger M, Weber W, Tei L, D'Alessandria C. Room Temperature Al 18 F Labeling of 2-Aminomethylpiperidine-Based Chelators for PET Imaging. ChemMedChem 2020; 15:284-292. [PMID: 31830368 DOI: 10.1002/cmdc.201900652] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/02/2019] [Indexed: 01/14/2023]
Abstract
Positron emission tomography (PET) is a non-invasive molecular imaging technology that is constantly expanding, with a high demand for specific antibody-derived imaging probes. The use of tracers based on temperature-sensitive molecules (i. e. Fab, svFab, nanobodies) is increasing and has led us to design a class of chelators based on the structure of 2-aminomethylpiperidine (AMP) with acetic and/or hydroxybenzyl pendant arms (2-AMPTA, NHB-2-AMPDA, and 2-AMPDA-HB), which were investigated as such for {Al18 F}2+ -core chelation efficiency. All the compounds were characterized by HPLC-MS analysis and NMR spectroscopy. The AlF-18 labeling reactions were performed under various conditions (pH/temperature), and the radiolabeled chelates were purified and characterized by radio-TLC and radio-HPLC. The stability of labeled chelates was investigated up to 240 min in human serum (HS), EDTA 5 mM, PBS and 0.9 % NaCl solutions. The in vivo stability of [Al18 F(2-AMPDA-HB)]- was assessed in healthy nude mice (n=6). Radiochemical yields between 55 % and 81 % were obtained at pH 5 and room temperature. High stability in HS was measured for [Al18 F(2-AMPDA-HB)]- , with 90 % of F-18 complexed after 120 min. High stability in vivo, rapid hepatobiliary and renal excretion, with low accumulation of free F-18 in bones were measured. Thus, this new Al18 F-chelator may have a great impact on immuno-PET radiopharmacy, by facilitating the development of new fluorine-18-labeled heat-sensitive biomolecules.
Collapse
Affiliation(s)
- Lisa Russelli
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Jonathan Martinelli
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121, Alessandria, Italy
| | - Francesco De Rose
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Sybille Reder
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Michael Herz
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Lorenzo Tei
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121, Alessandria, Italy
| | - Calogero D'Alessandria
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| |
Collapse
|
30
|
Ruiz-Bedoya CA, Gordon O, Mota F, Abhishek S, Tucker EW, Ordonez AA, Jain SK. Molecular Imaging of Diabetic Foot Infections: New Tools for Old Questions. Int J Mol Sci 2019; 20:E5984. [PMID: 31795077 PMCID: PMC6928969 DOI: 10.3390/ijms20235984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Diabetic foot infections (DFIs) are a common, complex, and costly medical problem with increasing prevalence. Diagnosing DFIs is a clinical challenge due to the poor specificity of the available methods to accurately determine the presence of infection in these patients. However, failure to perform an opportune diagnosis and provide optimal antibiotic therapy can lead to higher morbidity for the patient, unnecessary amputations, and increased healthcare costs. Novel developments in bacteria-specific molecular imaging can provide a non-invasive assessment of the infection site to support diagnosis, determine the extension and location of the infection, guide the selection of antibiotics, and monitor the response to treatment. This is a review of recent research in molecular imaging of infections in the context of DFI. We summarize different clinical and preclinical methods and the translational implications aimed to improve the care of patients with DFI.
Collapse
Affiliation(s)
- Camilo A. Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Oren Gordon
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Filipa Mota
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sudhanshu Abhishek
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth W. Tucker
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Pediatric Critical Care, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| | - Alvaro A. Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (C.A.R.-B.); (O.G.); (F.M.); (S.A.); (E.W.T.); (A.A.O.)
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
31
|
Toyohara J, Yamamoto H, Tago T. Searching for diagnostic properties of novel fluorine-18-labeled D-allose. Ann Nucl Med 2019; 33:855-865. [PMID: 31471865 DOI: 10.1007/s12149-019-01398-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/22/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Two fluorine-18-labeled analogues, 3-deoxy-3-[18F]fluoro-D-allose (3-[18F]FDA) and 6-deoxy-6-[18F]fluoro-D-allose (6-[18F]FDA), were synthesized and their potentials of diagnostic property were characterized. METHODS In vitro rat red blood cell (RBC) transport and phosphorylation by yeast hexokinase were evaluated in comparison with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). The rate of protein binding in pooled human serum was measured by an ultrafiltration method. In vivo metabolite analysis in mice was also performed. Biodistribution, urine excretion, and in vivo renal kinetics in mice were compared with 2-deoxy-2-[18F]fluorosorbitol ([18F]FDS). RESULTS Rat RBC uptake of 3- and 6-[18F]FDA (7.8 ± 2.5%ID and 10.2 ± 4.8%ID, respectively) was significantly lower than that of [18F]FDG (44.7 ± 8.7%ID). RBC uptake of 3-[18F]FDA was inhibited by D-glucose (30%) and cytochalasin B (40%), indicating the involvement of GLUT1-dependent transport. In contrast, 6-[18F]FDA transport was not inhibited by D-glucose and cytochalasin B. 3- and 6-[18F]FDA were not phosphorylated by yeast hexokinase under the conditions that result in 60% conversion of [18F]FDG into [18F]FDG-6-phosphate within 30 min. Serum protein binding of 3- and 6-[18F]FDA was negligible. Metabolic transformation of both tracers was not detected in plasma and urine at 30 min after injection. The highest tissue uptake of both tracers was observed in kidneys. Heart and brain uptake of both tracers was below blood levels throughout the biodistribution studies (until 120 min after injection). No significant uptake in the bone was observed, indicating the absence of de-fluorination in mice. In vivo PET imaging visualized rapid excretion of the administered 3- and 6-[18F]FDA from the kidneys, with minimal tracer accumulation in other organs. The urine excretion rate of 3-[18F]FDA was much lower than that of 6-[18F]FDA and [18F]FDS. CONCLUSIONS 3- and 6-[18F]FDA might be unsatisfactory for tumor imaging. In contrast, these tracers demonstrated high levels of kidney uptake and excretion, low serum protein binding, and high metabolic stability as preferable properties for renal imaging. Notably, the urine excretion rate and kidney uptake kinetics of 6-[18F]FDA were comparable with those of the potential renal imaging agent [18F]FDS. Further validation studies in animal models are required to confirm the feasibility of 6-[18F]FDA as a functional renal imaging agent.
Collapse
Affiliation(s)
- Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| | - Hiroyuki Yamamoto
- Department of Radiology and Nuclear Medicine, Akita Cerebrospinal and Cardiovascular Center, 6-10 Senshu-Kubota Machi, Akita, 010-0874, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| |
Collapse
|
32
|
Werner RA, Chen X, Lapa C, Koshino K, Rowe SP, Pomper MG, Javadi MS, Higuchi T. The next era of renal radionuclide imaging: novel PET radiotracers. Eur J Nucl Med Mol Imaging 2019; 46:1773-1786. [PMID: 31144061 PMCID: PMC6647203 DOI: 10.1007/s00259-019-04359-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/10/2019] [Indexed: 01/08/2023]
Abstract
Although single-photon-emitting radiotracers have long been the standard for renal functional molecular imaging, recent years have seen the development of positron emission tomography (PET) agents for this application. We provide an overview of renal radionuclide PET radiotracers, in particular focusing on novel 18F-labelled and 68Ga-labelled agents. Several reported PET imaging probes allow assessment of glomerular filtration rate, such as [68Ga]ethylenediaminetetraacetic acid ([68Ga]EDTA), [68Ga]IRDye800-tilmanocept and 2-deoxy-2-[18F]fluorosorbitol ([18F]FDS)). The diagnostic performance of [68Ga]EDTA has already been demonstrated in a clinical trial. [68Ga]IRDye800-tilmanocept shows receptor-mediated binding to glomerular mesangial cells, which in turn may allow the monitoring of progression of diabetic nephropathy. [18F]FDS shows excellent kidney extraction and excretion in rats and, as has been shown in the first study in humans. Further, due to its simple one-step radiosynthesis via the most frequently used PET radiotracer 2-deoxy-2-[18F]fluoro-D-glucose, [18F]FDS could be available at nearly every PET centre. A new PET radiotracer has also been introduced for the effective assessment of plasma flow in the kidneys: Re(CO)3-N-([18F]fluoroethyl)iminodiacetic acid (Re(CO)3([18F]FEDA)). This compound demonstrates similar pharmacokinetic properties to its 99mTc-labelled analogue [99mTc](CO)3(FEDA). Thus, if there is a shortage of molybdenum-99, Re(CO)3([18F]FEDA would allow direct comparison with previous studies with 99mTc. The PET radiotracers for renal imaging reviewed here allow thorough evaluation of kidney function, with the tremendous advantage of precise anatomical coregistration with simultaneously acquired CT images and rapid three-dimensional imaging capability.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany
- Comprehensive Heart Failure Center, University of Wuerzburg, Wuerzburg, Germany
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Xinyu Chen
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany
- Comprehensive Heart Failure Center, University of Wuerzburg, Wuerzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany
| | - Kazuhiro Koshino
- Department of Biomedical Imaging, National Cardiovascular and Cerebral Center, Suita, Japan
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mehrbod S Javadi
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany.
- Comprehensive Heart Failure Center, University of Wuerzburg, Wuerzburg, Germany.
- Department of Biomedical Imaging, National Cardiovascular and Cerebral Center, Suita, Japan.
- Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School, Okayama, Japan.
| |
Collapse
|
33
|
Abstract
The novel PET probe 2-deoxy-2-F-fluoro-D-sorbitol (F-FDS) has demonstrated favorable renal kinetics in animals. We aimed to elucidate its imaging properties in 2 human volunteers. F-FDS was produced by a simple 1-step reduction from F-FDG. On dynamic renal PET, the cortex was delineated and activity gradually transited in the parenchyma, followed by radiotracer excretion. No adverse effects were reported. Given the higher spatiotemporal resolution of PET relative to conventional scintigraphy, F-FDS PET offers a more thorough evaluation of human renal kinetics. Due to its simple production from F-FDG, F-FDS is virtually available at any PET facility with radiochemistry infrastructure.
Collapse
|
34
|
|
35
|
Ordonez AA, Jain SK. Pathogen-Specific Bacterial Imaging in Nuclear Medicine. Semin Nucl Med 2018. [DOI: 10.1053/j.semnuclmed.2017.11.003
expr 890398765 + 809902709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
36
|
Abstract
When serious infections are suspected, patients are often treated empirically with broad-spectrum antibiotics while awaiting results that provide information on the bacterial class and species causing the infection, as well as drug susceptibilities. For deep-seated infections, these traditional diagnostic techniques often rely on tissue biopsies to obtain clinical samples which can be expensive, dangerous, and has the potential of sampling bias. Moreover, these procedures and results can take several days and may not always provide reliable information. This combination of time and effort required for proper antibiotic selection has become a barrier leading to indiscriminate broad-spectrum antibiotic use. Exposure to nosocomial infections and indiscriminate use of broad-spectrum antibiotics are responsible for promoting bacterial drug-resistance leading to substantial morbidity and mortality, especially in hospitalized and immunosuppressed patients. Therefore, early diagnosis of infection and targeted antibiotic treatments are urgently needed to reduce morbidity and mortality caused by bacterial infections worldwide. Reliable pathogen-specific bacterial imaging techniques have the potential to provide early diagnosis and guide antibiotic treatments.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
37
|
Werner RA, Wakabayashi H, Chen X, Hirano M, Shinaji T, Lapa C, Rowe SP, Javadi MS, Higuchi T. Functional Renal Imaging with 2-Deoxy-2-18F-Fluorosorbitol PET in Rat Models of Renal Disorders. J Nucl Med 2017; 59:828-832. [DOI: 10.2967/jnumed.117.203828] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/27/2017] [Indexed: 11/16/2022] Open
|
38
|
Re: Increased 18F-2-Fluorodeoxysorbitol (18F-FDS) Activity in a Pituitary Spindle Cell Carcinoma. Clin Nucl Med 2017; 42:649. [PMID: 28319504 DOI: 10.1097/rlu.0000000000001644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Increased 18F-2- Fluorodeoxysorbitol (18F-FDS) Activity in a Pituitary Spindle Cell Carcinoma: Reply. Clin Nucl Med 2017; 42:649-650. [PMID: 28525462 DOI: 10.1097/rlu.0000000000001698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Zhu W, Yao S, Xing H, Zhang H, Tai YC, Zhang Y, Liu Y, Ma Y, Wu C, Wang H, Li Z, Wu Z, Zhu Z, Li F, Huo L. Biodistribution and Radiation Dosimetry of the Enterobacteriaceae-Specific Imaging Probe [(18)F]Fluorodeoxysorbitol Determined by PET/CT in Healthy Human Volunteers. Mol Imaging Biol 2017; 18:782-7. [PMID: 27020679 DOI: 10.1007/s11307-016-0946-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE [(18)F]fluorodeoxysorbitol ([(18)F]FDS) is the first radiopharmaceutical specific for a category of bacteria and has the potential to specifically detect Enterobacteriaceae infections. The purpose of this study was to testify the safety and investigate the biodistribution and radiation dosimetry of [(18)F]FDS in healthy human bodies. PROCEDURES Six healthy subjects were intravenously injected with 320-520 MBq [(18)F]FDS. On each subject, 21 whole-body emission scans and a brain scan were conducted at settled time points within the next 4 h. Residence time for each source organ was determined by multi-exponential regression. Absorbed doses for target organs and effective dose were calculated via OLINDA/EXM. RESULTS No adverse events due to [(18)F]FDS injection were observed in the study. The tracer was cleared rapidly from the blood pool through the urinary system. A small portion was cleared into the gut through the hepatobiliary system. The effective dose (ED) was estimated to be 0.021 ± 0.001 mSv/MBq. The organ receiving the highest absorbed dose was the urinary bladder wall (0.25 ± 0.03 mSv/MBq). CONCLUSIONS [(18)F]FDS is safe and well tolerated. The effective dose was comparable to that of other F-18 labeled radiotracers. [(18)F]FDS is suitable for human use from a radiation dosimetry perspective.
Collapse
Affiliation(s)
- Wenjia Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Shaobo Yao
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Haiqun Xing
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Yuan-Chuan Tai
- Department of Radiology, Washington University in St Louis, St Louis, MI, USA
| | - Yingqiang Zhang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yimin Liu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yanru Ma
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Chenxi Wu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Hongkai Wang
- Department of Biomedical Engineering, Dalian University of technology, Dalian, China
| | - Zibo Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhanhong Wu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, 1# Shuaifuyuan, Dongcheng District, Beijing, China.
| |
Collapse
|
41
|
Li J, Zheng H, Fodah R, Warawa JM, Ng CK. Validation of 2- 18F-Fluorodeoxysorbitol as a Potential Radiopharmaceutical for Imaging Bacterial Infection in the Lung. J Nucl Med 2017; 59:134-139. [PMID: 28848037 DOI: 10.2967/jnumed.117.195420] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/17/2017] [Indexed: 11/16/2022] Open
Abstract
2-18F-fluorodeoxysorbitol (18F-FDS) has been shown to be a promising agent with high selectivity and sensitivity in imaging bacterial infection. The objective of our study was to validate 18F-FDS as a potential radiopharmaceutical for imaging bacterial infection longitudinally in the lung. Methods: Albino C57 female mice were intratracheally inoculated with either live or dead Klebsiella pneumoniae to induce either lung infection or lung inflammation. One group of mice was imaged to monitor disease progression. PET/CT was performed on days 0, 1, 2, and 3 after inoculation using either 18F-FDS or 18F-FDG (n = 12 for each tracer). The other group was first screened by bioluminescent imaging (BLI) to select only mice with visible infection (region of interest > 108 ph/s) for PET/CT imaging with 18F-FDS (n = 12). For the inflammation group, 5 mice each were imaged with PET/CT using either 18F-FDS or 18F-FDG from days 1 to 4 after inoculation. Results: For studies of disease progression, BLI showed noticeable lung infection on day 2 after inoculation and significantly greater infection on day 3. Baseline imaging before inoculation showed no focal areas of lung consolidation on CT and low uptake in the lung for both PET radiotracers. On day 2, an area of lung consolidation was identified on CT, with a corresponding 2.5-fold increase over baseline for both PET radiotracers. On day 3, widespread areas of patchy lung consolidation were found on CT, with a drastic increase in uptake for both 18F-FDS and 18F-FDG (9.2 and 3.9). PET and BLI studies showed a marginal correlation between 18F-FDG uptake and colony-forming units (r = 0.63) but a much better correlation for 18F-FDS (r = 0.85). The uptake ratio of infected lung over inflamed lung was 8.5 and 1.7 for 18F-FDS and 18F-FDG on day 3. Conclusion: Uptake of both 18F-FDS and 18F-FDG in infected lung could be used to track the degree of bacterial infection measured by BLI, with a minimum detection limit of 107 bacteria. 18F-FDS, however, is more specific than 18F-FDG in differentiating K. pneumoniae lung infection from lung inflammation.
Collapse
Affiliation(s)
- Junling Li
- University of Louisville School of Medicine, Louisville, Kentucky
| | - Huaiyu Zheng
- University of Louisville School of Medicine, Louisville, Kentucky
| | - Ramy Fodah
- University of Louisville School of Medicine, Louisville, Kentucky
| | | | - Chin K Ng
- University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
42
|
Dutta J, Naicker T, Ebenhan T, Kruger HG, Arvidsson PI, Govender T. Synthetic approaches to radiochemical probes for imaging of bacterial infections. Eur J Med Chem 2017; 133:287-308. [DOI: 10.1016/j.ejmech.2017.03.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 02/08/2023]
|
43
|
Increased 18F-2-Fluorodeoxysorbitol (18F-FDS) Activity in a Pituitary Spindle Cell Carcinoma. Clin Nucl Med 2017; 41:953-955. [PMID: 27764039 DOI: 10.1097/rlu.0000000000001391] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A 33-year-old woman presented with progressive visual loss. An MRI examination revealed a pituitary lesion which was suspicious of abscess. FDG and F-2-fluorodeoxysorbitol (F-FDS) PET were performed to differentiate malignancy from inflammation. FDS is a new tracer with potential in differentiating inflammatory tissues based on results from animal studies. However, intense tracer uptake was found in the pituitary in both PET studies. The lesion was proved to be spindle cell carcinoma following biopsy.
Collapse
|
44
|
Neuroimaging. IMAGING INFECTIONS 2017. [PMCID: PMC7123586 DOI: 10.1007/978-3-319-54592-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Imaging of infection in the CNS has been handled using cross-sectional imaging for more than two decades now resulting in a large array of descriptive diagnostic criteria, capable, in most circumstances of narrowing the differential diagnosis, detecting life-threatening complications and establishing baseline for assessment of treatment response. Limitations however exist, and in many circumstances, both cross-sectional imaging and nonspecific molecular imaging, such as 18F-FDG, fail to establish a diagnosis. The availability of pathogen-specific imaging agents/ligands would have a great effect on the management of patients with CNS infection. Besides early diagnosis, avoidance of diagnostic brain biopsies can have significant effect on the mortality and morbidity of patients.
Collapse
|
45
|
Ordonez AA, Weinstein EA, Bambarger LE, Saini V, Chang YS, DeMarco VP, Klunk MH, Urbanowski ME, Moulton KL, Murawski AM, Pokkali S, Kalinda AS, Jain SK. A Systematic Approach for Developing Bacteria-Specific Imaging Tracers. J Nucl Med 2016; 58:144-150. [PMID: 27635025 DOI: 10.2967/jnumed.116.181792] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022] Open
Abstract
The modern patient is increasingly susceptible to bacterial infections including those due to multidrug-resistant organisms (MDROs). Noninvasive whole-body analysis with pathogen-specific imaging technologies can significantly improve patient outcomes by rapidly identifying a source of infection and monitoring the response to treatment, but no such technology exists clinically. METHODS We systematically screened 961 random radiolabeled molecules in silico as substrates for essential metabolic pathways in bacteria, followed by in vitro uptake in representative bacteria-Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, and mycobacteria. Fluorine-labeled analogs, that could be developed as PET-based imaging tracers, were evaluated in a murine myositis model. RESULTS We identified 3 novel, nontoxic molecules demonstrating selective bacterial uptake: para-aminobenzoic acid (PABA), with uptake in all representative bacteria including Mycobacterium tuberculosis; mannitol, with selective uptake in S. aureus and E. coli; and sorbitol, accumulating only in E. coli None accumulated in mammalian cells or heat-killed bacteria, suggesting metabolism-derived specificity. In addition to an extended bacterial panel of laboratory strains, all 3 molecules rapidly accumulated in respective clinical isolates of interest including MDROs such as methicillin-resistant S. aureus, extended-spectrum β-lactamase-producing, and carbapenem-resistant Enterobacteriaceae. In a murine myositis model, fluorine-labeled analogs of all 3 molecules could rapidly detect and differentiate infection sites from sterile inflammation in mice (P = 0.03). Finally, 2-deoxy-2-[F-18]fluoro-d-sorbitol (18F-FDS) can be easily synthesized from 18F-FDG. PET, with 18F-FDS synthesized using current good manufacturing practice, could rapidly differentiate true infection from sterile inflammation to selectively localize E. coli infection in mice. CONCLUSION We have developed a systematic approach that exploits unique biochemical pathways in bacteria to develop novel pathogen-specific imaging tracers. These tracers have significant potential for clinical translation to specifically detect and localize a broad range of bacteria, including MDROs.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward A Weinstein
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Lauren E Bambarger
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vikram Saini
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yong S Chang
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vincent P DeMarco
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mariah H Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael E Urbanowski
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kimberly L Moulton
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Allison M Murawski
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Supriya Pokkali
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alvin S Kalinda
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland .,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
46
|
Wakabayashi H, Werner RA, Hayakawa N, Javadi MS, Xinyu C, Herrmann K, Rowe SP, Lapa C, Higuchi T. Initial Preclinical Evaluation of 18F-Fluorodeoxysorbitol PET as a Novel Functional Renal Imaging Agent. J Nucl Med 2016; 57:1625-1628. [PMID: 27013700 DOI: 10.2967/jnumed.116.172718] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/22/2016] [Indexed: 01/04/2023] Open
Abstract
Accurate assessment of kidney function plays an essential role for optimal clinical decision making in a variety of diseases. The major intrinsic advantages of PET are superior spatial and temporal resolutions for quantitative tomographic renal imaging. 2-deoxy-2-18F-fluorodeoxysorbitol (18F-FDS) is an analog of sorbitol that is reported to be freely filtered at the renal glomerulus without reabsorption at the tubule. Furthermore, it can be synthesized via simple reduction of widely available 18F-FDG. We tested the feasibility of 18F-FDS renal PET imaging in rats. METHODS The systemic and renal distribution of 18F-FDS were determined by dynamic 35-min PET imaging (15 frames × 8 s, 26 frames × 30 s, 20 frames × 60 s) with a dedicated small-animal PET system and postmortem tissue counting in healthy rats. Distribution of coinjected 99mTc-diethylenetriaminepentaacetic acid (DTPA) was also estimated as a reference. Plasma binding and in vivo stability of 18F-FDS were determined. RESULTS In vivo PET imaging visualized rapid excretion of the administrated 18F-FDS from both kidneys, with minimal tracer accumulation in other organs. Initial cortical tracer uptake followed by visualization of the collecting system could be observed with high contrast. Split-function renography curves were successfully obtained in healthy rats (the time of maximal concentration [Tmax] right [R] = 2.8 ± 1.2 min, Tmax left [L] = 2.9 ± 1.5 min, the time of half maximal concentration [T1/2max] R = 8.8 ± 3.7 min, T1/2max L = 11.1 ± 4.9 min). Postmortem tissue counting of 18F-FDS confirmed the high kidney extraction (kidney activities at 10, 30, and 60 min after tracer injection [percentage injected dose per gram]: 1.8 ± 0.7, 1.2 ± 0.1, and 0.5 ± 0.2, respectively) in a degree comparable to 99mTc-DTPA (2.5 ± 1.0, 1.5 ± 0.2, and 0.8 ± 0.3, respectively). Plasma protein binding of 18F-FDS was low (<0.1%), and metabolic transformation was not detected in serum and urine. CONCLUSION In rat experiments, 18F-FDS demonstrated high kidney extraction and excretion, low plasma protein binding, and high metabolic stability as preferable properties for renal imaging. These preliminary results warrant further confirmatory studies in large animal models and clinical studies as a novel functional renal imaging agent, given the advantages of PET technology and broad tracer availability.
Collapse
Affiliation(s)
| | - Rudolf A Werner
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany; and
| | - Nobuyuki Hayakawa
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Mehrbod S Javadi
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Chen Xinyu
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany; and
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Constantin Lapa
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany; and
| |
Collapse
|
47
|
Abstract
The most commonly utilized PET radionuclide is fluorine-18 ((18)F) because of its convenient half-life and excellent imaging properties. In this review, we present the first analysis of patents issued for radiotracers labeled with fluorine-18 (between 2009 and 2015), and provide perspective on current trends and future directions in PET radiotracer development.
Collapse
|
48
|
Yao S, Xing H, Zhu W, Wu Z, Zhang Y, Ma Y, Liu Y, Huo L, Zhu Z, Li Z, Li F. Infection Imaging With (18)F-FDS and First-in-Human Evaluation. Nucl Med Biol 2015; 43:206-14. [PMID: 26924501 DOI: 10.1016/j.nucmedbio.2015.11.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/07/2015] [Accepted: 11/27/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE The noninvasive imaging of bacterial infections is critical in order to reduce mortality and morbidity caused by these diseases. The recently reported (18)F-FDS ((18)F-2-fluorodeoxy sorbitol) as a PET (positron emission tomography) tracer can be used to image Enterobacteriaceae-specific infections and provides a potential alternative to this problem compared with other probes for imaging infections. In this study, automatic synthesis, validation of (18)F-FDS and a first-in-human study were performed and discussed. METHODS A multifunctional synthesis module was employed for the radiosynthesis of (18)F-FDG ((18)F-2-fluorodeoxy glucose) and (18)F-FDS starting from (18)F ion using two-pot three-step fully automated reactions. The behavior of (18)F-FDS as an in vivo imaging probe for infections was evaluated in an Escherichia coli mouse infection model. The first detailed pharmacokinetic and biodistribution parameters were obtained from healthy human volunteers. RESULTS The uptake of (18)F-FDS in an E. coli mouse-myositis infection model was easily differentiated from other organs and normal muscle. Intensive lesion uptake declined after antibiotic treatment. In the pilot human study, no adverse effects due to (18)F-FDS were observed up to 24 h post-injection. The radiotracer was rapidly cleared from the circulation and excreted mainly through the urinary system. CONCLUSION We conclude that (18)F-FDS PET holds great potential for appropriate and effective for the imaging of bacterial infections in vivo. These preliminary results indicate that further clinical studies are warranted.
Collapse
Affiliation(s)
- Shaobo Yao
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Haiqun Xing
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Zhanhong Wu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Yingqiang Zhang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Yanru Ma
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Yimin Liu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Zibo Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
49
|
Abstract
Enterobacteriaceae, a type of highly virulent Gram-negative bacteria, can be imaged in vivo in animals via positron emission tomography with (18)F-sorbitol (Weinstein et al., this issue).
Collapse
Affiliation(s)
- Xiaojian Wang
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
50
|
Weinstein EA, Ordonez AA, DeMarco VP, Murawski AM, Pokkali S, MacDonald EM, Klunk M, Mease RC, Pomper MG, Jain SK. Imaging Enterobacteriaceae infection in vivo with 18F-fluorodeoxysorbitol positron emission tomography. Sci Transl Med 2015; 6:259ra146. [PMID: 25338757 DOI: 10.1126/scitranslmed.3009815] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The Enterobacteriaceae are a family of rod-shaped Gram-negative bacteria that normally inhabit the gastrointestinal tract and are the most common cause of Gram-negative bacterial infections in humans. In addition to causing serious multidrug-resistant, hospital-acquired infections, a number of Enterobacteriaceae species are also recognized as biothreat pathogens. As a consequence, new tools are urgently needed to specifically identify and localize infections due to Enterobacteriaceae and to monitor antimicrobial efficacy. In this report, we used commercially available 2-[(18)F]-fluorodeoxyglucose ((18)F-FDG) to produce 2-[(18)F]-fluorodeoxysorbitol ((18)F-FDS), a radioactive probe for Enterobacteriaceae, in 30 min. (18)F-FDS selectively accumulated in Enterobacteriaceae, but not in Gram-positive bacteria or healthy mammalian or cancer cells in vitro. In a murine myositis model, (18)F-FDS positron emission tomography (PET) rapidly differentiated true infection from sterile inflammation with a limit of detection of 6.2 ± 0.2 log10 colony-forming units (CFU) for Escherichia coli. Our findings were extended to models of mixed Gram-positive and Gram-negative thigh co-infections, brain infection, Klebsiella pneumonia, and mice undergoing immunosuppressive chemotherapy. This technique rapidly and specifically localized infections due to Enterobacteriaceae, providing a three-dimensional holistic view within the animal. Last, (18)F-FDS PET monitored the efficacy of antimicrobial treatment, demonstrating a PET signal proportionate to the bacterial burden. Therapeutic failures associated with multidrug-resistant, extended-spectrum β-lactamase (ESBL)-producing E. coli infections were detected in real time. Together, these data show that (18)F-FDS is a candidate imaging probe for translation to human clinical cases of known or suspected infections owing to Enterobacteriaceae.
Collapse
Affiliation(s)
- Edward A Weinstein
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Vincent P DeMarco
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Allison M Murawski
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Supriya Pokkali
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth M MacDonald
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mariah Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ronnie C Mease
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Martin G Pomper
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|