1
|
Jiang Y, Wang Q, Feng J, Yin G, Han P, Ruan Q, Zhang J. A novel Al 18F-labelled NOTA-modified ubiquicidin 29-41 derivative as a bacterial infection PET imaging agent. Eur J Med Chem 2025; 289:117482. [PMID: 40058182 DOI: 10.1016/j.ejmech.2025.117482] [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: 01/16/2025] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
The antimicrobial peptide ubiquicidin 29-41 (TGRAKRRMQYNRR) is a potential target for detecting bacterial infection. A novel UBI 29-41 derivative modified with 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) on the amino side of lysine was synthesized and radiolabelled with Al18F, named [18F]AlF-NOTA-UBI 29-41. The novel PET tracer maintained good in vitro stability in saline at room temperature and mouse serum at 37 °C. In vitro bacterial binding experiments indicated that the tracer specifically bound to Staphylococcus aureus. A significant difference in the uptake of [18F]AlF-NOTA-UBI 29-41 between infected muscle and inflamed muscle was observed in biodistribution. A PET imaging study in mouse models with bacterial infection and sterile inflammation showed apparent accumulation at the infection site, suggesting that the complex is a potential PET tracer for distinguishing bacterial infection from sterile inflammation.
Collapse
Affiliation(s)
- Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China; Department of Nuclear Technology and Application, China Institute of Atomic Energy, Beijing, 102413, China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Peiwen Han
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China; Key Laboratory of Beam Technology of the Ministry of Education, School of Physics and Astronomy, Beijing Normal University, Beijing, 100875, China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
2
|
Mizutani A, Kondo A, Muranaka Y, Momose Y, Nishiyama Y, Sato K, Kobayashi M, Kawai K. Application of [ 99mTc]Tc-GSA in the diagnosis of Staphylococcus aureus infections. Nucl Med Biol 2025; 146-147:109021. [PMID: 40306183 DOI: 10.1016/j.nucmedbio.2025.109021] [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: 02/05/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Staphylococcus aureus can cause a variety of conditions such as bacteremia, sepsis, toxic shock syndrome, pneumonia, and infective endocarditis; therefore, a rapid and accurate diagnosis should be made to pinpoint the site of infection. This study aimed to use the existing nuclear medicine tracer [99mTc]Tc-dimercaptosuccinic acid galactosyl human serum albumin ([99mTc]Tc-GSA) as a simple technique for the early diagnosis of S. aureus infection. METHODS The in vitro studies evaluated the accumulation of [99mTc]Tc-GSA in S. aureus. In addition, the effect of metabolic and vital activity and inhibition of asialoglycoprotein receptors on [99mTc]Tc-GSA accumulation were evaluated. In vivo studies were performed on the biodistribution and imaging of [99mTc]Tc-GSA in the S. aureus SR3637 mouse thigh infection model. RESULTS In vitro studies have confirmed that [99mTc]Tc-GSA accumulates to the same extent as 2-deoxy-2-[18F]fluoro-d-glucose, and it was thought that [99mTc]Tc-GSA binds to the receptors that recognize the saccharide molecules and glycan chains expressed in S. aureus. In addition, in the distribution and imaging of [99mTc]Tc-GSA, it was confirmed that the latter accumulates at the infection site and shows a clear contrast with the non-infected site. CONCLUSION The application of [99mTc]Tc-GSA to the imaging diagnosis of S. aureus infection is expected to non-invasively detect the localization of S. aureus in real time, pinpoint the site of infection and determine the number of viable bacteria, and help in the selection of optimal therapeutic agents and treatments.
Collapse
Affiliation(s)
- Asuka Mizutani
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan
| | - Ami Kondo
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan
| | - Yuka Muranaka
- Department of Radiological Technology, Faculty of Health Science, Juntendo University, 2-1-1 Hongo, Bunkyo-ku 113-8421, Tokyo, Japan
| | - Yuma Momose
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan
| | - Yuri Nishiyama
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan; Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka 561-0825, Osaka, Japan
| | - Kakeru Sato
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan; Radiological Center, University of Fukui Hospital, 23-3 Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun 910-1193, Fukui, Japan
| | - Masato Kobayashi
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan
| | - Keiichi Kawai
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan; Biomedical Imaging Research Center, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun 910-1193, Fukui, Japan.
| |
Collapse
|
3
|
Dvorakova Bendova K, Krasulova K, Neuzilova B, Popper M, Mlynarcik P, Hajduova K, Novy Z, Hajduch M, Petrik M. Positron Emission Tomography Imaging of Acinetobacter baumannii Infection: Comparison of Gallium-68 Labeled Siderophores. ACS Infect Dis 2025; 11:917-928. [PMID: 40099411 PMCID: PMC11997986 DOI: 10.1021/acsinfecdis.4c00946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Acinetobacter baumannii (AB) is an opportunistic pathogen with growing clinical relevance due to its increasing level of antimicrobial resistance in the last few decades. In the event of an AB hospital outbreak, fast detection and localization of the pathogen is crucial, to prevent its further spread. However, contemporary diagnostic tools do not always meet the requirements for rapid and accurate diagnosis. For this reason, we report here the possibility of using gallium-68 labeled siderophores, bacterial iron chelators, for positron emission tomography imaging of AB infections. In our study, we radiolabeled several siderophores and tested their in vitro uptake in AB cultures. Based on the results and the in vitro properties of studied siderophores, we selected two of them for further in vivo testing in infectious models. Both selected siderophores, ferrioxamine E and ferrirubin, showed promising in vitro characteristics. In vivo, we observed rapid pharmacokinetics and no excessive accumulation in organs other than the excretory organs in normal mice. We demonstrated that the radiolabeled siderophores accumulate in AB-infected tissue in three animal models: a murine model of myositis, a murine model of dorsal wound infection and a rat model of pneumonia. These results suggest that both siderophores radiolabeled with Ga-68 could be used for PET imaging of AB infection.
Collapse
Affiliation(s)
- Katerina Dvorakova Bendova
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
| | - Kristyna Krasulova
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
| | - Barbora Neuzilova
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
| | - Miroslav Popper
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
| | - Patrik Mlynarcik
- Department
of Microbiology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, 775 15 Olomouc, Czech Republic
| | - Katarina Hajduova
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
| | - Zbynek Novy
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
- Czech
Advanced Technology and Research Institute, Palacký University, 779 00 Olomouc, Czech Republic
| | - Marian Hajduch
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
- Laboratory
of Experimental Medicine, University Hospital, 779 00 Olomouc, Czech Republic
| | - Milos Petrik
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00 Olomouc, Czech Republic
- Laboratory
of Experimental Medicine, University Hospital, 779 00 Olomouc, Czech Republic
- Czech
Advanced Technology and Research Institute, Palacký University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
4
|
Yu Y, Cui T, Liu C, Yang W, Zhang B. Tunable Hierarchically Porous Gadolinium-Based Metal-Organic Frameworks for Bacteria-Targeting Magnetic Resonance Imaging and In Situ Anti-Bacterial Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415209. [PMID: 39976077 PMCID: PMC12005816 DOI: 10.1002/advs.202415209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Currently, there are no non-invasive tools to accurately diagnose deep surgical site bacterial infections before they cause significant anatomical damage in the clinic. An urgent need exists for bacteria-targeting bifunctional probes for the detection of deep bacterial infections and precise in situ treatment. Herein, the bacteria-targeting 1-borono-3,5-benzenedicarboxylic acid (BBDC) ligand and paramagnetic Gd3 + into one single metal-organic frameworks (MOFs) are integrated, synergistically realizing bacteria-specific magnetic resonance imaging (MRI) diagnosis and MRI-guided antibacterial treatment. Molecular simulations and nitrogen adsorption-desorption experiments demonstrate that a hierarchical porous structure can be constructed by tuning the Gd3 + /BBDC ratio, which endows the Gd-BBDC1.25 MOFs with an impressive longitudinal proton relaxivity of 15.81 mM-1 s-1. In particular, the bacteria-targeting boronic acid group in BBDC remained intact during the MOF synthesis, ensuring that Gd-BBDC1.25 MOFs have a unique combination of high sensitivity and specificity for bacteria. Through an in situ reduction reaction, silver nanoparticles (Ag NPs)-modified Gd-BBDC1.25 MOFs to form Ag@Gd-BBDC1.25, an interfacial Schottky heterojunction nanozyme, which enhances their peroxidase (POD)-catalyze activity. Furthermore, it is demonstrated that the bacteria-targeting Ag@Gd-BBDC1.25 bifunctional probe can image as few as 105 colony-forming units (cfu) in vivo and effectively eradicate the bacteria in situ.
Collapse
Affiliation(s)
- Youyi Yu
- Department of RadiologyTongji HospitalShanghai Frontiers Science Center of Nanocatalytic Medicinethe Institute for Biomedical Engineering & Nano ScienceSchool of MedicineTongji UniversityShanghai200065China
| | - Tianming Cui
- Shanghai Research Institute for Intelligent Autonomous SystemsTongji UniversityShanghai200065China
| | - Chang Liu
- Department of RadiologyTongji HospitalShanghai Frontiers Science Center of Nanocatalytic Medicinethe Institute for Biomedical Engineering & Nano ScienceSchool of MedicineTongji UniversityShanghai200065China
| | - Weitao Yang
- Department of RadiologyTongji HospitalShanghai Frontiers Science Center of Nanocatalytic Medicinethe Institute for Biomedical Engineering & Nano ScienceSchool of MedicineTongji UniversityShanghai200065China
| | - Bingbo Zhang
- Department of RadiologyTongji HospitalShanghai Frontiers Science Center of Nanocatalytic Medicinethe Institute for Biomedical Engineering & Nano ScienceSchool of MedicineTongji UniversityShanghai200065China
| |
Collapse
|
5
|
Jiang Y, Wang Q, Yin G, Feng J, Ruan Q, Han P, Zhang J. Development of Novel 99mTc-Labeled Hydrazinoicotinamide-Modified Ubiquicidin 29-41 Complexes with Improved Target-to-Nontarget Ratios for Bacterial Infection Imaging. ACS Pharmacol Transl Sci 2025; 8:470-483. [PMID: 39974636 PMCID: PMC11833726 DOI: 10.1021/acsptsci.4c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
To develop novel 99mTc-labeled ubiquicidin 29-41 derivatives for bacterial infection SPECT imaging aiming at achieving a high target-to-nontarget ratio and lower nontarget organ uptake, a novel 6-hydrazinoicotinamide (HYNIC) ubiquicidin 29-41 derivative (HYNIC-UBI 29-41) was designed and synthesized. It was then radiolabeled with ternary ligands, including TPPTS, PDA, 2,6-PDA, NIC, ISONIC, PSA, 4-PSA, and PES, to obtain eight 99mTc-labeled HYNIC-UBI 29-41 complexes. All the complexes demonstrated hydrophilicity, exhibited good in vitro stability, and specifically bound Staphylococcus aureus in vitro. Biodistribution studies in mice with bacterial infection demonstrated that [99mTc]Tc-tricine/TPPTS-HYNIC-UBI 29-41 resulted in increased abscess-to-muscle and abscess-to-blood ratios as well as decreased nontarget organ uptake. Furthermore, it was able to distinguish between bacterial infection and sterile inflammation. Single-photon emission computed tomography (SPECT) imaging studies in mice with bacterial infection revealed visible accumulation at the site of infection, indicating that [99mTc]Tc-tricine/TPPTS-HYNIC-UBI 29-41 is a potential radiotracer for imaging bacterial infection.
Collapse
Affiliation(s)
- Yuhao Jiang
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Qianna Wang
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Guangxing Yin
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Junhong Feng
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
- Department
of Nuclear Technology and Application, China
Institute of Atomic Energy, Beijing 102413, China
| | - Qing Ruan
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
- Key
Laboratory
of Beam Technology of the Ministry of Education, School of Physics
and Astronomy, Beijing Normal University, Beijing 100875, China
| | - Peiwen Han
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Junbo Zhang
- Key Laboratory
of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
6
|
González-Arjona M, Sobrino G, Cussó L, Guembe M, Calle D, Díaz Crespo F, Bouza E, Muñoz P, Desco M, Salinas B. 99mTc-DTPA-Collagen Radiotracer for the Noninvasive Detection of Infective Endocarditis. ACS Infect Dis 2025; 11:121-130. [PMID: 39645608 PMCID: PMC11731287 DOI: 10.1021/acsinfecdis.4c00460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 12/09/2024]
Abstract
Infective endocarditis (IE) represents a significant concern among hospital-acquired infections, frequently caused by the Gram-positive bacterium Staphylococcus aureus. Nuclear imaging is emerging as a noninvasive and precise diagnostic tool. However, the gold standard radiotracer [18F]-FDG cannot distinguish between infection and inflammation, resulting in false positives. Based on the presence of collagen-binding proteins in the cell wall of S. aureus, we propose the radiolabeling of collagen for its evaluation in IE animal models by single-photon emission computed tomography (SPECT) imaging. We radiolabeled rat tail collagen I using DTPA chelator and [99mTc]NaTcO4. Selectivity was evaluated in vitro using 3 Gram-positive bacteria, 1 Gram-negative bacteria and 1 yeast. In vivo SPECT/computed tomography (CT) imaging was conducted on 8 SD rat models of IE and 8 sterile sham model as controls. Ex vivo biodistribution and autoradiography were performed following imaging. Diagnosis of IE was confirmed through microbiological studies and H&E histopathology. [99mTc]-DTPA-Collagen was synthesized successfully with a yield of 42.86 ± 6.35%, a purity of 95.84 ± 1.85% and a stability higher than 90% after 50 h postincubation. In vitro uptake demonstrated the selectivity for Gram-positive bacteria (63.85 ± 15.15%). Ex vivo analysis confirmed hepato-splenic excretion. In vivo SPECT/CT imaging revealed highly localized uptake within the aortic valve with a sensitivity of 62.5% and specificity of 87.5%. We successfully synthesized and characterized a new SPECT radiotracer based on [99mTc]Tc-radiolabeled collagen. In vitro studies demonstrated the selectivity of the radiotracer for Gram-positive bacteria. In vivo SPECT/CT-based assessment in an IE model confirmed the potential of this approach to detect active IE.
Collapse
Affiliation(s)
- Mario González-Arjona
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Gorka Sobrino
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Lorena Cussó
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid 28029, Spain
| | - María Guembe
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Servicio
de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain
| | - Daniel Calle
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Francisco Díaz Crespo
- Servicio
de Anatomía Patológica, Hospital
General Universitario Gregorio Marañón, Madrid 28007, Spain
| | - Emilio Bouza
- Servicio
de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain
- Departamento
de Medicina, Facultad de Medicina, Universidad
Complutense de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Enfermedades Respiratorias
(CIBERES), Madrid 28029, Spain
| | - Patricia Muñoz
- Servicio
de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid 28007, Spain
- Departamento
de Medicina, Facultad de Medicina, Universidad
Complutense de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Enfermedades Respiratorias
(CIBERES), Madrid 28029, Spain
| | - Manuel Desco
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid 28029, Spain
- Departamento
de Bioingeniería, Universidad Carlos
III de Madrid, Madrid 28911, Spain
| | - Beatriz Salinas
- Unidad
de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Instituto
de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Unidad
de Imagen Avanzada, Centro Nacional de Investigaciones
Cardiovasculares (CNIC), Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid 28029, Spain
- Departamento
de Bioingeniería, Universidad Carlos
III de Madrid, Madrid 28911, Spain
| |
Collapse
|
7
|
Pollard-Kerning AC, Li K, Li Y, Ahn SH, Wang M, Akoglu M, Bravo E, DelloRusso F, Akula HK, Qu W, Meimetis L, Schlyer DJ, Komatsu DE, Tonge PJ. Preclinical Positron Emission Tomography (PET) of Prosthetic Joint Infection Using a Nitro-Prodrug of 2-[ 18F]F- p-Aminobenzoic Acid ([ 18F]F-PABA). ACS Infect Dis 2024; 10:3765-3774. [PMID: 39432749 DOI: 10.1021/acsinfecdis.4c00075] [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: 10/23/2024]
Abstract
Deep-seated bacterial infections are difficult to detect and diagnose due to the lack of specific clinical imaging modalities. Therefore, the bacteria-specific positron emission tomography radiotracer 2-[18F]fluoro-4-nitrobenzoic acid ([18F]FNB) was developed, which is reduced to 2-[18F]fluoro-4-aminobenzoic acid ([18F]F-PABA) by bacterial nitroreductases and has improved pharmacokinetics compared to the parent compound. PET imaging demonstrated that the uptake of 2-[18F]fluoro-4-nitrobenzoic acid in a clinically relevant Staphylococcus aureus prosthetic joint infection model was up to ∼4-fold higher in the infected joint compared to the contralateral joint. 2-[18F]Fluoro-4-nitrobenzoic acid was also able to distinguish infection from inflammation in a surgical inflammation model. Based on the mouse radiation dosimetry results, the calculated effective dose of 2-[18F]fluoro-4-nitrobenzoic acid was well below the whole-body radiation dose limit established by the Food and Drug Administration for humans. In addition, no treatment-related microscopic changes in organ histopathology were observed in a mouse acute toxicity study. Overall, these data suggest that 2-[18F]fluoro-4-nitrobenzoic acid is a specific and effective imaging agent for noninvasively diagnosing prosthetic joint infections.
Collapse
Affiliation(s)
- Alyssa C Pollard-Kerning
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
- Chronus Pharmaceuticals Inc., Long Island High Technology Incubator, 25 Health Sciences Drive, Stony Brook, New York 11790-3350, United States
| | - Kaixuan Li
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Yong Li
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
- Chronus Pharmaceuticals Inc., Long Island High Technology Incubator, 25 Health Sciences Drive, Stony Brook, New York 11790-3350, United States
| | - Shin Hye Ahn
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
- Chronus Pharmaceuticals Inc., Long Island High Technology Incubator, 25 Health Sciences Drive, Stony Brook, New York 11790-3350, United States
| | - Mingqian Wang
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Melike Akoglu
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Eduardo Bravo
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Francesca DelloRusso
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Hari K Akula
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York 11794-8101, United States
| | - Wenchao Qu
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York 11794-8101, United States
| | - Labros Meimetis
- Chronus Pharmaceuticals Inc., Long Island High Technology Incubator, 25 Health Sciences Drive, Stony Brook, New York 11790-3350, United States
- Department of Radiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York 11794-8460, United States
| | - David J Schlyer
- Isotope Research and Production, Brookhaven National Lab, Upton, New York 11973-5000, United States
| | - David E Komatsu
- Department of Orthopedics, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York 11794-8181, United States
| | - Peter J Tonge
- Center for Advanced Study of Drug Action and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
8
|
Saleem SM, Jabbar T, Imran MB, Noureen A, Sherazi TA, Afzal MS, Rab Nawaz HZ, Ramadan MF, Alkahtani AM, Alsuwat MA, Almubarak HA, Momenah MA, Naqvi SAR. Radiosynthesis and Preclinical Evaluation of [ 99mTc]Tc-Tigecycline Radiopharmaceutical to Diagnose Bacterial Infections. Pharmaceuticals (Basel) 2024; 17:1283. [PMID: 39458924 PMCID: PMC11510260 DOI: 10.3390/ph17101283] [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: 08/23/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES As a primary source of mortality and disability, bacterial infections continue to develop a severe threat to humanity. Nuclear medicine imaging (NMI) is known for its promising potential to diagnose deep-seated bacterial infections. This work aims to develop a new technetium-99m (99mTc) labeled tigecycline radiopharmaceutical as an infection imaging agent. METHODS Reduced 99mTc was used to make a coordinate complex with tigecycline at pH 7.7-7.9 at room temperature. Instantaneous thin-layer chromatography impregnated with silica gel (ITLC-SG) and ray detector equipped high-performance liquid chromatography (ray-HPLC) was performed to access the radiolabeling yield and radiochemical purity (RCP). RESULTS More than 91% labeling efficiency was achieved after 25 min of mild shaking of the reaction mixture. The radiolabeled complex was found intact up to 4 h in saline. Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) infection-induced rats were used to record the biodistribution of the radiopharmaceutical and its target specificity; 2 h' post-injection biodistribution revealed a 2.39 ± 0.29 target/non-target (T/NT) ratio in the E. coli infection-induced animal model, while a 2.9 ± 0.31 T/NT value was recorded in the S. aureus bacterial infection-induced animal model. [99mTc]Tc-tigecycline scintigraphy was performed in healthy rabbits using a single photon emission computed tomography (SPECT) camera. Scintigrams showed normal kidney perfusion and excretion into the bladder. CONCLUSION In conclusion, the newly developed [99mTc]Tc-tigecycline radiopharmaceutical could be considered to diagnose broad-spectrum bacterial infections.
Collapse
Affiliation(s)
- Syeda Marab Saleem
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38040, Pakistan
| | - Tania Jabbar
- Punjab Institute of Nuclear Medicine, Faisalabad 38040, Pakistan
| | | | - Asma Noureen
- Department of Zoology, Ghazi University, Dera Ghazi Khan 03222, Pakistan
| | - Tauqir A. Sherazi
- Department of Chemistry, COMSAT University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | | | - Hafiza Zahra Rab Nawaz
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38040, Pakistan
| | - Mohamed Fawzy Ramadan
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Abdullah M. Alkahtani
- Department of Microbiology & Clinical Parasitology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Meshari A. Alsuwat
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Hassan Ali Almubarak
- Assistant Professor Nuclear Medicine, Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, Abha 61421, Saudi Arabia
| | - Maha Abdullah Momenah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Syed Ali Raza Naqvi
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38040, Pakistan
| |
Collapse
|
9
|
Gouws CA, Naicker T, de la Torre BG, Albericio F, Duvenhage J, Kruger HG, Marjanovic-Painter B, Mdanda S, Zeevaart JR, Ebenhan T, Govender T. 68Ga Radiolabeling of NODASA-Functionalized Phage Display-Derived Peptides for Prospective Assessment as Tuberculosis-Specific PET Radiotracers. J Labelled Comp Radiopharm 2024; 67:360-374. [PMID: 39118205 DOI: 10.1002/jlcr.4120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024]
Abstract
This research presents the development of positron emission tomography (PET) radiotracers for detecting Mycobacterium tuberculosis (MTB) for the diagnosis and monitoring of tuberculosis. Two phage display-derived peptides with proven selective binding to MTB were identified for development into PET radiopharmaceuticals: H8 (linear peptide) and PH1 (cyclic peptide). We sought to functionalize H8/PH1 with NODASA, a bifunctional chelator that allows complexation of PET-compatible radiometals such as gallium-68. Herein, we report on the chelator functionalization, optimized radiosynthesis, and assessment of the radiopharmaceutical properties of [68Ga]Ga-NODASA-H8 and [68Ga]Ga-NODASA-PH1. Robust radiolabeling was achieved using the established routine method, indicating consistent production of a radiochemically pure product (RCP ≥ 99.6%). For respective [68Ga]Ga-NODASA-H8 and [68Ga]Ga-NODASA-PH1, relatively high levels of decay-corrected radiochemical yield (91.2% ± 2.3%, 86.7% ± 4.0%) and apparent molar activity (Am, 3.9 ± 0.8 and 34.0 ± 5.3 GBq/μmol) were reliably achieved within 42 min, suitable for imaging purposes. Notably, [68Ga]Ga-NODASA-PH1 remained stable in blood plasma for up to 2 h, while [68Ga]Ga-NODASA-H8 degraded within 30 min. For both 68Ga peptides, minimal whole-blood cell binding and plasma protein binding were observed, indicating a favorable pharmaceutical behavior. [68Ga]Ga-NODASA-PH1 is a promising candidate for further in vitro/in vivo evaluation as a tuberculosis-specific infection imaging agent.
Collapse
Affiliation(s)
- Christiaan A Gouws
- Catalysis and Peptide Research Unit, School of Health Sciences and School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Tricia Naicker
- Catalysis and Peptide Research Unit, School of Health Sciences and School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | | | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Janie Duvenhage
- Pre-clinical Imaging Facility (PCIF), Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, School of Health Sciences and School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | | | - Sipho Mdanda
- Pre-clinical Imaging Facility (PCIF), Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
| | - Jan R Zeevaart
- Pre-clinical Imaging Facility (PCIF), Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
- Radiochemistry, the South African Nuclear Energy Corporation (Necsa) SOC Ltd, Pelindaba, South Africa
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa
| | - Thomas Ebenhan
- Pre-clinical Imaging Facility (PCIF), Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa
| | | |
Collapse
|
10
|
Li K, Collado JT, Marden JA, Pollard AC, Guo S, Tonge PJ, Qu W. Biological Evaluation of d-[ 18F]Fluoroalanine and d-[ 18F]Fluoroalanine- d3 as Positron Emission Tomography Imaging Tracers for Bacterial Infection. J Med Chem 2024; 67:13975-13984. [PMID: 39082959 PMCID: PMC11342404 DOI: 10.1021/acs.jmedchem.4c00783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
d-Amino acids such as d-alanine are substrates for bacterial peptidoglycan biosynthesis and are selectively taken up by bacteria and not mammalian cells. Consequently, d-amino acid metabolism is an attractive target for antibiotic discovery and the development of bacteria-specific imaging agents. d-Fluoroalanine and the deuterium-labeled analogue fludalanine (MK641) were originally explored as antibiotics by Merck but failed in clinical trials due to unaccepted toxicity. Herein, we synthesized a fluorine-18 labeled d-fluoroalanine, d-3-[18F]fluoroalanine (d-[18F]FAla), and its deuterated analogue, d-3-[18F]fluoroalanine-d3 (d-[18F]FAla-d3), and evaluated their capability to image bacterial infection. Both d-[18F]FAla and d-[18F]FAla-d3 can accumulate up to 0.64-0.78% ID/cc in the infectious area at 15 min postinjection. Despite the reduction of in vivo defluorination not being observed for deuterated 18F-labeled d-fluoroalanine, these radiolabeled d-alanine analogues were able to differentiate bacterial infection from sterile inflammation in a soft-tissue model of S. aureus infection.
Collapse
Affiliation(s)
- Kaixuan Li
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Jinnette Tolentino Collado
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Jocelyn A. Marden
- Department of Psychiatry and Behavioral Health, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| | - Alyssa C. Pollard
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Shuwen Guo
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
| | - Peter J. Tonge
- Center for Advanced Study of Drug Action, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Radiology, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
- Stony Brook Cancer Center, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| | - Wenchao Qu
- Department of Chemistry, John S. Toll Drive, Stony Brook University, Stony Brook, NY 11794-3400, United States
- Department of Psychiatry and Behavioral Health, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
- PET Research Core, Stony Brook Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, United States
| |
Collapse
|
11
|
Akter A, Firth G, Darwesh AMF, Cooper MS, Chuljerm H, Cilibrizzi A, Blower PJ, Hider RC, Lyons O, Schelenz S, Mehra V, Abbate V. [ 68Ga]Ga-Schizokinen, a Potential Radiotracer for Selective Bacterial Infection Imaging. ACS Infect Dis 2024; 10:2615-2622. [PMID: 39012184 PMCID: PMC11320569 DOI: 10.1021/acsinfecdis.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024]
Abstract
Gallium-68-labeled siderophores as radiotracers have gained interest for the development of in situ infection-specific imaging diagnostics. Here, we report radiolabeling, in vitro screening, and in vivo pharmacokinetics (PK) of gallium-68-labeled schizokinen ([68Ga]Ga-SKN) as a new potential radiotracer for imaging bacterial infections. We radiolabeled SKN with ≥95% radiochemical purity. Our in vitro studies demonstrated its hydrophilic characteristics, neutral pH stability, and short-term stability in human serum and toward transchelation. In vitro uptake of [68Ga]Ga-SKN by Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and S. epidermidis, but no uptake by Candida glabrata, C. albicans, or Aspergillus fumigatus, demonstrated its specificity to bacterial species. Whole-body [68Ga]Ga-SKN positron emission tomography (PET) combined with computerized tomography (CT) in healthy mice showed rapid renal excretion with no or minimal organ uptake. The subsequent ex vivo biodistribution resembled this fast PK with rapid renal excretion with minimal blood retention and no major organ uptake and showed some dissociation of the tracer in the urine after 60 min postinjection. These findings warrant further evaluation of [68Ga]Ga-SKN as a bacteria-specific radiotracer for infection imaging.
Collapse
Affiliation(s)
- Asma Akter
- Institute
of Pharmaceutical Science, Faculty of Life Science and Medicine, King’s College London, London SE1 9NH, United Kingdom
| | - George Firth
- School
of Biomedical Engineering and Imaging Sciences, Faculty of Life Science
and Medicine, King’s College London, London SE1 7EH, United Kingdom
| | - Afnan M. F. Darwesh
- School
of Biomedical Engineering and Imaging Sciences, Faculty of Life Science
and Medicine, King’s College London, London SE1 7EH, United Kingdom
- Department
of Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Margaret S. Cooper
- School
of Biomedical Engineering and Imaging Sciences, Faculty of Life Science
and Medicine, King’s College London, London SE1 7EH, United Kingdom
| | - Hataichanok Chuljerm
- Institute
of Pharmaceutical Science, Faculty of Life Science and Medicine, King’s College London, London SE1 9NH, United Kingdom
- School
of Health Sciences Research, Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Agostino Cilibrizzi
- Institute
of Pharmaceutical Science, Faculty of Life Science and Medicine, King’s College London, London SE1 9NH, United Kingdom
| | - Philip J. Blower
- School
of Biomedical Engineering and Imaging Sciences, Faculty of Life Science
and Medicine, King’s College London, London SE1 7EH, United Kingdom
| | - Robert C. Hider
- Institute
of Pharmaceutical Science, Faculty of Life Science and Medicine, King’s College London, London SE1 9NH, United Kingdom
| | - Oliver Lyons
- Department
of Surgery, University of Otago, Christchurch 8013, New Zealand
| | - Silke Schelenz
- Department
of Microbiology, Kings College Hospital
NHS Foundation Trust, London SE5 9RS, United
Kingdom
| | - Varun Mehra
- Department
of Hematology, King’s College Hospital
NHS Foundation Trust, London SE5 9RS, United
Kingdom
| | - Vincenzo Abbate
- Institute
of Pharmaceutical Science, Faculty of Life Science and Medicine, King’s College London, London SE1 9NH, United Kingdom
| |
Collapse
|
12
|
Betts HM, Luckett JC, Hill PJ. Pilot Evaluation of S-(3-[ 18F]Fluoropropyl)-D-Homocysteine and O-(2-[ 18F]Fluoroethyl)-D-Tyrosine as Bacteria-Specific Radiotracers for PET Imaging of Infection. Mol Imaging Biol 2024; 26:704-713. [PMID: 38942967 PMCID: PMC11282134 DOI: 10.1007/s11307-024-01929-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/30/2024]
Abstract
PURPOSE There is currently no ideal radiotracer for imaging bacterial infections. Radiolabelled D-amino acids are promising candidates because they are actively incorporated into the peptidoglycan of the bacterial cell wall, a structural feature which is absent in human cells. This work describes fluorine-18 labelled analogues of D-tyrosine and D-methionine, O-(2-[18F]fluoroethyl)-D-tyrosine (D-[18F]FET) and S-(3-[18F]fluoropropyl)-D-homocysteine (D-[18F]FPHCys), and their pilot evaluation studies as potential radiotracers for imaging bacterial infection. PROCEDURES D-[18F]FET and D-[18F]FPHCys were prepared in classical fluorination-deprotection reactions, and their uptake in Staphylococcus aureus and Pseudomonas aeruginosa was evaluated over 2 h. Heat killed bacteria were used as controls. A clinically-relevant foreign body model of S. aureus infection was established in Balb/c mice, as well as a sterile foreign body to mimic inflammation. The ex vivo biodistribution of D-[18F]FPHCys in the infected and inflamed mice was evaluated after 1 h, by dissection and gamma counting. The uptake was compared to that of [18F]FDG. RESULTS In vitro uptake of both D-[18F]FET and D-[18F]FPHCys was specific to live bacteria. Uptake was higher in S. aureus than in P. aeruginosa for both radiotracers, and of the two, higher for D-[18F]FPHCys than D-[18F]FET. Blocking experiments with non-radioactive D-[19F]FPHCys confirmed specificity of uptake. In vivo, D-[18F]FPHCys had greater accumulation in S. aureus infection compared with sterile inflammation, which was statistically significant. As anticipated, [18F]FDG showed no significant difference in uptake between infection and inflammation. CONCLUSIONS D-[18F]FPHCys uptake was higher in infected tissues than inflammation, and represents a fluorine-18 labelled D-AA with potential to detect a S. aureus reference strain (Xen29) in vivo. Additional studies are needed to evaluate uptake of this radiotracer in clinical isolates.
Collapse
Affiliation(s)
- Helen M Betts
- Department of Nuclear Medicine, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
- School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Jeni C Luckett
- School of Life Sciences, University of Nottingham, Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Philip J Hill
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington, LE17 5RD, UK
| |
Collapse
|
13
|
Li Y, Zhou Y, Du Y, Gao P, Yang L, Wang W. In vivo Labeling and Intravital Imaging of Bacterial Infection using a Near-infrared Fluorescent D-Amino Acid Probe. Chembiochem 2024; 25:e202400283. [PMID: 38715148 DOI: 10.1002/cbic.202400283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/28/2024] [Indexed: 06/27/2024]
Abstract
Bacterial infections still pose a severe threat to public health, necessitating novel tools for real-time analysis of microbial behaviors in living organisms. While genetically engineered strains with fluorescent or luminescent reporters are commonly used in tracking bacteria, their in vivo uses are often limited. Here, we report a near-infrared fluorescent D-amino acid (FDAA) probe, Cy7ADA, for in situ labeling and intravital imaging of bacterial infections in mice. Cy7ADA probe effectively labels various bacteria in vitro and pathogenic Staphylococcus aureus in mice after intraperitoneal injection. Because of Cy7's high tissue penetration and the quick excretion of free probes via urine, real-time visualization of the pathogens in a liver abscess model via intravital confocal microscopy is achieved. The biodistributions, including their intracellular localization within Kupffer cells, are revealed. Monitoring bacterial responses to antibiotics also demonstrates Cy7ADA's capability to reflect the bacterial load dynamics within the host. Furthermore, Cy7ADA facilitates three-dimensional pathogen imaging in tissue-cleared liver samples, showcasing its potential for studying the biogeography of microbes in different organs. Integrating near-infrared FDAA probes with intravital microscopy holds promise for wide applications in studying bacterial infections in vivo.
Collapse
Affiliation(s)
- Yixuan Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Yingjun Zhou
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Microbiome Center, School of Life Sciences, Fudan University, Shanghai, China
| | - Yahui Du
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Liqun Yang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Wei Wang
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Microbiome Center, School of Life Sciences, Fudan University, Shanghai, China
- Beijing National Laboratory for Molecular Sciences, Beijing, China
| |
Collapse
|
14
|
Kahts M, Summers B, Gutta A, Pilloy W, Ebenhan T. Recently developed radiopharmaceuticals for bacterial infection imaging. EJNMMI Radiopharm Chem 2024; 9:49. [PMID: 38896373 PMCID: PMC11187059 DOI: 10.1186/s41181-024-00279-7] [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: 04/18/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Infection remains a major cause of morbidity and mortality, regardless of advances in antimicrobial therapy and improved knowledge of microorganisms. With the major global threat posed by antimicrobial resistance, fast and accurate diagnosis of infections, and the reliable identification of intractable infection, are becoming more crucial for effective treatment and the application of antibiotic stewardship. Molecular imaging with the use of nuclear medicine allows early detection and localisation of infection and inflammatory processes, as well as accurate monitoring of treatment response. There has been a continuous search for more specific radiopharmaceuticals to be utilised for infection imaging. This review summarises the most prominent discoveries in specifically bacterial infection imaging agents over the last five years, since 2019. MAIN BODY Some promising new radiopharmaceuticals evaluated in patient studies are reported here, including radiolabelled bacterial siderophores like [68Ga]Ga-DFO-B, radiolabelled antimicrobial peptide/peptide fragments like [68Ga]Ga-NOTA-UBI29-41, and agents that target bacterial synthesis pathways (folic acid and peptidoglycan) like [11C]para-aminobenzoic acid and D-methyl-[11C]-methionine, with clinical trials underway for [18F]fluorodeoxy-sorbitol, as well as for 11C- and 18F-labelled trimethoprim. CONCLUSION It is evident that a great deal of effort has gone into the development of new radiopharmaceuticals for infection imaging over the last few years, with remarkable progress in preclinical investigations. However, translation to clinical trials, and eventually clinical Nuclear Medicine practice, is apparently slow. It is the authors' opinion that a more structured and harmonised preclinical setting and well-designed clinical investigations are the key to reliably evaluate the true potential of the newly proposed infection imaging agents.
Collapse
Affiliation(s)
- Maryke Kahts
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University, Ga-Rankuwa, 0208, South Africa.
| | - Beverley Summers
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University, Ga-Rankuwa, 0208, South Africa
| | - Aadil Gutta
- Nuclear Medicine Department, Dr George Mukhari Academic Hospital, Ga-Rankuwa, 0208, South Africa
- School of Medicine, Sefako Makgatho Health Sciences University, Ga-Rankuwa, 0208, South Africa
| | - Wilfrid Pilloy
- Nuclear Medicine Department, Dr George Mukhari Academic Hospital, Ga-Rankuwa, 0208, South Africa
| | - Thomas Ebenhan
- Nuclear Medicine Department and Nuclear Medicine Research Infrastructure, University of Pretoria, Pretoria, 0001, South Africa
| |
Collapse
|
15
|
Dai D, Yu J, Gou W, Yang S, Li Y, Wang Z, Yang Z, Huang T, Li P, Zhu T, Hou W, Zhao Y, Xu W, Li Y. Novel CDK19-Targeted Radiotracers: A Potential Design Strategy to Improve the Pharmacokinetics and Tumor Uptake. J Med Chem 2024; 67:6726-6737. [PMID: 38570733 DOI: 10.1021/acs.jmedchem.4c00281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Cyclin-dependent kinase 19 (CDK19) is overexpressed in prostate cancer, making it an attractive target for both imaging and therapy. Since little is known about the optimized approach for radioligands of nuclear proteins, linker optimization strategies were used to improve pharmacokinetics and tumor absorption, including the adjustment of the length, flexibility/rigidity, and hydrophilicity/lipophilicity of linkers. Molecular docking was conducted for virtual screening and followed by IC50 determination. Both BALB/c mice and P-16 xenografts were used for tissue distribution and PET/CT imaging. The ligand 68Ga-10c demonstrated high absorption in tumor 5 min after injection and sustains long-term imaging within 3 h. Furthermore, 68Ga-10c exhibited slow clearance within the tumor and was predominantly metabolized in both the liver and kidneys, showing the potential to alleviate metabolic pressure and enhance tissue safety. Therefore, the linker optimization strategy is well suited for CDK19 and provides a reference for the radioactive ligands of other nuclear targets.
Collapse
Affiliation(s)
- Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin 300060, China
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Jiang Yu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wenfeng Gou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Shuangmeng Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yanli Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Ziyang Wang
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Zhao Yang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin 300060, China
| | - Ting Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Panfeng Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wenbin Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin 300060, China
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
16
|
Nogueira SA, Barboza MRFFD, Bezerra RP, Cabeza JM, Dell'Aquila AM, Santos DDCB, Yamaga LYI, Osawa A. Antimicrobial peptide for bacterial infection imaging: first case reported in Brazil. EINSTEIN-SAO PAULO 2023; 21:eRC0621. [PMID: 38055555 DOI: 10.31744/einstein_journal/2023rc0621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/14/2023] [Indexed: 12/08/2023] Open
Abstract
Molecular imaging markers can be used to differentiate between infection and aseptic inflammation, determine the severity of infection, and monitor treatment responses. One of these markers is ubiquicidin(29-41) (UBI), a cationic peptide fragment that binds to the bacterial membrane wall and is labeled with gallium-68 (68Ga), a positron emitter radioisotope. The use of UBI in positron emission tomography (PET)/computed tomography (CT) for improved detection of lesions has been receiving considerable attention recently. Herein, we report the first case of 68Ga-UBI PET/CT performed in Brazil. The patient was a 39-year-old woman referred for a scan to confirm a clinical suspicion of chronic osteomyelitis of her fractured left tibia. PET images revealed radiotracer uptake near the posterior contour of the tibial fracture focus and the fixation plate, in the soft tissue around the distal half of the tibia, and in the non-consolidated fracture of the left distal fibula. Surgery for local cleaning was performed, and culture of a specimen collected from the surgical site confirmed the presence of Staphylococcus aureus. In the present case, 68Ga-UBI PET/CT, a non-invasive imaging modality, identified the infection foci in vivo, indicating its potential for clinical use.
Collapse
Affiliation(s)
- Solange Amorim Nogueira
- Department of Radiology and Diagnostic Imaging, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Rosemeire Pereira Bezerra
- Department of Radiology and Diagnostic Imaging, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Jorge Mejia Cabeza
- Department of Radiology and Diagnostic Imaging, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Adriana Macedo Dell'Aquila
- Department of Medicine and Infectious Diseases, Hospital do Servidor Público Estadual de São Paulo, São Paulo, SP, Brazil
| | | | - Lilian Yuri Itaya Yamaga
- Department of Radiology and Diagnostic Imaging, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Akemi Osawa
- Department of Radiology and Diagnostic Imaging, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| |
Collapse
|
17
|
Alberto S, Ordonez AA, Arjun C, Aulakh GK, Beziere N, Dadachova E, Ebenhan T, Granados U, Korde A, Jalilian A, Lestari W, Mukherjee A, Petrik M, Sakr T, Cuevas CLS, Welling MM, Zeevaart JR, Jain SK, Wilson DM. The Development and Validation of Radiopharmaceuticals Targeting Bacterial Infection. J Nucl Med 2023; 64:1676-1682. [PMID: 37770110 PMCID: PMC10626374 DOI: 10.2967/jnumed.123.265906] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
The International Atomic Energy Agency organized a technical meeting at its headquarters in Vienna, Austria, in 2022 that included 17 experts representing 12 countries, whose research spanned the development and use of radiolabeled agents for imaging infection. The meeting focused largely on bacterial pathogens. The group discussed and evaluated the advantages and disadvantages of several radiopharmaceuticals, as well as the science driving various imaging approaches. The main objective was to understand why few infection-targeted radiotracers are used in clinical practice despite the urgent need to better characterize bacterial infections. This article summarizes the resulting consensus, at least among the included scientists and countries, on the current status of radiopharmaceutical development for infection imaging. Also included are opinions and recommendations regarding current research standards in this area. This and future International Atomic Energy Agency-sponsored collaborations will advance the goal of providing the medical community with innovative, practical tools for the specific image-based diagnosis of infection.
Collapse
Affiliation(s)
- Signore Alberto
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, University of Rome "Sapienza," Rome, Italy
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chanda Arjun
- Radiopharmaceutical Program, Board of Radiation and Isotope Technology, Mumbai, India
| | - Gurpreet Kaur Aulakh
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Nicolas Beziere
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Thomas Ebenhan
- Nuclear Medicine, University of Pretoria, and Radiochemistry, Applied Radiation, South African Nuclear Energy Corporation, Pelindaba, South Africa
| | - Ulises Granados
- Department of Nuclear Medicine, Hospital Internacional de Colombia-Fundación Cardiovascular de Colombia, Piedecuesta, Colombia
| | - Aruna Korde
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Amirreza Jalilian
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Wening Lestari
- National Nuclear Energy Agency, South Tangerang, Indonesia
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Milos Petrik
- Institute of Molecular and Translational Medicine and Czech Advanced Technology and Research Institute, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Tamer Sakr
- Radioactive Isotopes and Generator Department, Hot Labs Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | | | - Mick M Welling
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Jan Rijn Zeevaart
- Nuclear Medicine, University of Pretoria, and Radiochemistry, Applied Radiation, South African Nuclear Energy Corporation, Pelindaba, South Africa
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| |
Collapse
|
18
|
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: 1.5] [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
|
19
|
Kleynhans J, Sathekge MM, Ebenhan T. Preclinical Research Highlighting Contemporary Targeting Mechanisms of Radiolabelled Compounds for PET Based Infection Imaging. Semin Nucl Med 2023; 53:630-643. [PMID: 37012169 DOI: 10.1053/j.semnuclmed.2023.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 04/04/2023]
Abstract
It is important to constantly monitor developments in the preclinical imaging arena of infection. Firstly, novel radiopharmaceuticals with the correct characteristics must be identified to funnel into the clinic. Secondly, it must be evaluated if enough innovative research is being done and adequate resources are geared towards the development of radiopharmaceuticals that could feed into the Nuclear Medicine Clinic in the near future. It is proposed that the ideal infection imaging agent will involve PET combined with CT but more ideally MRI. The radiopharmaceuticals currently presented in preclinical literature have a wide selection of vectors and targets. Ionic formulations of PET-radionuclides such 64CuCl2 and 68GaCl2 are evaluated for bacterial infection imaging. Many small molecule based radiopharmaceuticals are being investigated with the most prominent targets being cell wall synthesis, maltodextrin transport (such as [18F]F-maltotriose), siderophores (bacterial and fungal infections), the folate synthesis pathway (such as [18F]F-PABA) and protein synthesis (radiolabelled puromycin). Mycobacterial specific antibiotics, antifungals and antiviral agents are also under investigation as infection imaging agents. Peptide based radiopharmaceuticals are developed for bacterial, fungal and viral infections. The radiopharmaceutical development could even react quickly enough on a pandemic to develop a SARS-CoV-2 imaging agent in a timely fashion ([64Cu]Cu-NOTA-EK1). New immuno-PET agents for the imaging of viruses have recently been published, specifically for HIV persistence but also for SARS-CoV2. A very promising antifungal immuno-PET agent (hJ5F) is also considered. Future technologies could include the application of aptamers and bacteriophages and even going as far as the design of theranostic infection. Another possibility would be the application of nanobodies for immuno-PET applications. Standardization and optimization of the preclinical evaluation of radiopharmaceuticals could enhance clinical translation and reduce time spent in pursuing less than optimal candidates.
Collapse
Affiliation(s)
- Janke Kleynhans
- Department of Pharmaceutical and Pharmacological sciences, Radiopharmaceutical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mike Machaba Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa; Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Thomas Ebenhan
- Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, South Africa; Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
20
|
Aguilera-Correa JJ, Salinas B, González-Arjona M, de Pablo D, Muñoz P, Bouza E, Fernández Aceñero MJ, Esteban J, Desco M, Cussó L. Positron Emission Tomography-Computed Tomography and Magnetic Resonance Imaging Assessments in a Mouse Model of Implant-Related Bone and Joint Staphylococcus aureus Infection. Microbiol Spectr 2023; 11:e0454022. [PMID: 37010409 PMCID: PMC10269916 DOI: 10.1128/spectrum.04540-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/04/2023] [Indexed: 04/04/2023] Open
Abstract
Osteomyelitis is an infection of the bone, associated with an inflammatory process. Imaging plays an important role in establishing the diagnosis and the most appropriate patient management. However, data are lacking regarding the use of preclinical molecular imaging techniques to assess osteomyelitis progression in experimental models. This study aimed to compare structural and molecular imaging to assess disease progression in a mouse model of implant-related bone and joint infections caused by Staphylococcus aureus. In SWISS mice, the right femur was implanted with a resorbable filament impregnated with S. aureus (infected group, n = 10) or sterile culture medium (uninfected group, n = 6). Eight animals (5 infected, 3 uninfected) were analyzed with magnetic resonance imaging (MRI) at 1, 2, and 3 weeks postintervention, and 8 mice were analyzed with [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET)-computed tomography (CT) at 48 h and at 1, 2, and 3 weeks postintervention. In infected animals, CT showed bone lesion progression, mainly in the distal epiphysis, although some uninfected animals presented evident bone sequestra at 3 weeks. MRI showed a lesion in the articular area that persisted for 3 weeks in infected animals. This lesion was smaller and less evident in the uninfected group. At 48 h postintervention, FDG-PET showed higher joint uptake in the infected group than in the uninfected group (P = 0.025). Over time, the difference between groups increased. These results indicate that FDG-PET imaging was much more sensitive than MRI and CT for differentiating between infection and inflammation at early stages. FDG-PET clearly distinguished between infection and postsurgical bone healing (in uninfected animals) from 48 h to 3 weeks after implantation. IMPORTANCE Our results encourage future investigations on the utility of the model for testing different therapeutic procedures for osteomyelitis.
Collapse
Affiliation(s)
- J. J. Aguilera-Correa
- Departamento de Química en Ciencias Farmacéuticas. Universidad Complutense de Madrid, Madrid, Spain
- CIBERINFEC-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - B. Salinas
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III. Madrid, Spain
| | - M. González-Arjona
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - D. de Pablo
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Fundación para la Investigación Biomédica HCSC, Madrid, Spain
| | - P. Muñoz
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- CIBER Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - E. Bouza
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- CIBER Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - M. J. Fernández Aceñero
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Fundación para la Investigación Biomédica HCSC, Madrid, Spain
| | - J. Esteban
- CIBERINFEC-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Microbiology Department, IIS-Fundacion Jimenez Diaz, UAM, Madrid, Spain
| | - M. Desco
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III. Madrid, Spain
| | - L. Cussó
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III. Madrid, Spain
| |
Collapse
|
21
|
Li L, Liu M, Deng S, Zhu X, Song Y, Song E. Enzyme-Triggered Transforming of Assembly Peptide-Modified Magnetic Resonance-Tuned Probe for Highly Sensitive Imaging of Bacterial Infection In Vivo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208249. [PMID: 36929641 DOI: 10.1002/smll.202208249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/12/2023] [Indexed: 06/18/2023]
Abstract
Confirming bacterial infection at an early stage and distinguishing between sterile inflammation and bacterial infection is still highly needed for efficient treatment. Here, in situ highly sensitive magnetic resonance imaging (MRI) bacterial infection in vivo based on a peptide-modified magnetic resonance tuning (MRET) probe (MPD-1) that responds to matrix metallopeptidase 2 (MMP-2) highly expressed in bacteria-infected microenvironments is achieved. MPD-1 is an assembly of magnetic nanoparticle (MNP) bearing with gadolinium ion (Gd3+ ) modified MMP-2-cleavable self-assembled peptide (P1 ) and bacteria-targeting peptide (P), and it shows T2 -weighted signal due to the assemble of MNP and MRET ON phenomenon between MNP assembly and Gd3+ . Once MPD-1 accumulates at the bacterially infected site, P1 included in MPD-1 is cleaved explicitly by MMP-2, which triggers the T2 contrast agent of MPD-1 to disassemble into the monomer of MNP, leading the recovery of T1 -weighted signal. Simultaneously, Gd3+ detaches from MNP, further enhancing the T1 -weighted signal due to MRET OFF. The sensitive MRI of Staphylococcus aureus (low to 104 CFU) at the myositis site and accurate differentiation between sterile inflammation and bacterial infection based on the proposed MPD-1 probe suggests that this novel probe would be a promising candidate for efficiently detecting bacterial infection in vivo.
Collapse
Affiliation(s)
- Linyao Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| | - Maojuan Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| | - Siyu Deng
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| | - Xiaokang Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, CAS, Beijing, 100085, P. R. China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
22
|
Singh SB, Bhandari S, Siwakoti S, Bhatta R, Raynor WY, Werner TJ, Alavi A, Hess S, Revheim ME. Is Imaging Bacteria with PET a Realistic Option or an Illusion? Diagnostics (Basel) 2023; 13:diagnostics13071231. [PMID: 37046449 PMCID: PMC10093025 DOI: 10.3390/diagnostics13071231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
The application of [18F]-fluorodeoxyglucose ([18F]FDG) as a radiotracer to detect sites of inflammation (either due to bacterial infection or primary inflammation) has led to exploring the role of PET in visualizing bacteria directly at sites of infection. However, the results from such efforts are controversial and inconclusive so far. We aimed to assess the limitations of PET as an effective modality in the diagnosis of bacterial infections. Inflammation due to bacterial infections can be visualized by using [18F]FDG-PET. However, the non-specificity of [18F]FDG makes it undesirable to visualize bacteria as the underlying cause of inflammation. Hence, more specific radiotracers that possibly bind to or accumulate in bacteria-specific receptors or enzymes are being explored. Several radiotracers, including 2-deoxy-2-[18F]fluorosorbitol ([18F]FDS), 6-[18F]-fluoromaltose, [11C]para-aminobenzoic acid ([11C]PABA), radiolabeled trimethoprim (11C-TMP) and its analog fluoropropyl-trimethoprim (18F-FPTMP), other radiolabeled sugars, and antimicrobial drugs have been used to image microorganisms. Unfortunately, no progress has been made in translating the results to routine human use; feasibility and other factors have constrained their success in clinical settings. In the current article, we discuss the limitations of direct bacterial visualization with PET tracers, but emphasize the important role of [18F]FDG-PET as the only option for detecting evidence of infection.
Collapse
Affiliation(s)
- Shashi B Singh
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Sadikshya Bhandari
- Kathmandu University School of Medical Sciences, Dhulikhel Hospital, Dhulikhel 45200, Nepal
| | - Shisir Siwakoti
- Kathmandu University School of Medical Sciences, Dhulikhel Hospital, Dhulikhel 45200, Nepal
| | - Rabi Bhatta
- Universal College of Medical Sciences, Bhairahawa 32900, Nepal
| | - William Y Raynor
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, 1 Robert Wood Johnson Place, MEB #404, New Brunswick, NJ 08901, USA
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Soren Hess
- Department of Radiology and Nuclear Medicine, Hospital Southwest Jutland, 6700 Esbjerg, Denmark
- Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, 5230 Odense, Denmark
| | - Mona-Elisabeth Revheim
- The Intervention Center, Division of Technology and Innovation, Oslo University Hospital, 0424 Oslo, Norway
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, 0424 Oslo, Norway
- Norway and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0315 Oslo, Norway
| |
Collapse
|
23
|
Wang Y, Lin C, Zhang Z, Shen L, Zou B. Directed Nickel-Catalyzed Selective Arylhydroxylation of Unactivated Alkenes under Air. Org Lett 2023; 25:2172-2177. [PMID: 36946921 DOI: 10.1021/acs.orglett.3c00085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
An expeditious and novel nickel-catalyzed selective arylhydroxylation of unactivated alkenes with arylboronic acids was developed. This protocol is compatible with β,γ- and γ,δ-alkene amides, including traditionally challenging internal alkenes, to provide important β-arylethylalcohol scaffolds. The free hydroxyl group in the final product could be smoothly further transformed into other functional groups. Control experiments indicated that the oxygen atom of the hydroxyl group in the product is derived from the oxygen in the air.
Collapse
Affiliation(s)
- Yihua Wang
- College of Chemistry and Chemical Engineering, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Cong Lin
- College of Chemistry and Chemical Engineering, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Zongxu Zhang
- College of Chemistry and Chemical Engineering, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Liang Shen
- Jiangxi Engineering Laboratory of Waterborne Coatings, College of Chemistry and Chemical Engineering, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Boya Zou
- College of Chemistry and Chemical Engineering, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| |
Collapse
|
24
|
Liu F, Chen S, Zou Y, Jiao Y, Tang Y. A simple and efficient fluorescent labeling method in Staphylococcus aureus for real-time tracking of invasive bacteria. Front Microbiol 2023; 14:1128638. [PMID: 36846783 PMCID: PMC9950555 DOI: 10.3389/fmicb.2023.1128638] [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: 12/21/2022] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
Bacterial fluorescent labeling is a powerful tool for the diagnosis and treatment of bacterial infections. Here, we present a simple and efficient labeling strategy for Staphylococcus aureus. Intracellular labeling of bacteria was achieved by heat shock using Cyanine 5.5 (Cy5.5) near-infrared-I dyes in S. aureus (Cy5.5@S. aureus). Several key factors, such as Cy5.5 concentration and labeling time, were systematically evaluated. Further, the cytotoxicity of Cy5.5 and the stability of Cy5.5@S. aureus was evaluated by flow cytometry, inverted fluorescence microscopy, and transmission electron microscopy. In addition, Cy5.5@S. aureus were used to explore the phagocytic behavior of RAW264.7 macrophages. These results proved that Cy5.5@S. aureus had a uniform fluorescence intensity and high luminance; additionally, our method had no significant adverse effects on S. aureus compared to unlabeled S. aureus infections. Our method provides researchers with a useful option for analyzing the behavior of S. aureus as an infectious agent. This technique can be broadly applied to study host cell-bacteria interactions at the molecular level, and to in vivo tracing of bacterial infections.
Collapse
Affiliation(s)
- Fei Liu
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Sijie Chen
- Department of Nursing, The 940th Hospital of Joint Logistic Support Force of People’s Liberation Army (PLA), Lanzhou, China
| | - Yingxin Zou
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Yong Jiao
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Ying Tang
- Naval Medical Center, Naval Medical University, Shanghai, China,*Correspondence: Ying Tang,
| |
Collapse
|
25
|
Akter A, Lyons O, Mehra V, Isenman H, Abbate V. Radiometal chelators for infection diagnostics. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 2:1058388. [PMID: 37388440 PMCID: PMC7614707 DOI: 10.3389/fnume.2022.1058388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Infection of native tissues or implanted devices is common, but clinical diagnosis is frequently difficult and currently available noninvasive tests perform poorly. Immunocompromised individuals (for example transplant recipients, or those with cancer) are at increased risk. No imaging test in clinical use can specifically identify infection, or accurately differentiate bacterial from fungal infections. Commonly used [18F]fluorodeoxyglucose (18FDG) positron emission computed tomography (PET/CT) is sensitive for infection, but limited by poor specificity because increased glucose uptake may also indicate inflammation or malignancy. Furthermore, this tracer provides no indication of the type of infective agent (bacterial, fungal, or parasitic). Imaging tools that directly and specifically target microbial pathogens are highly desirable to improve noninvasive infection diagnosis and localization. A growing field of research is exploring the utility of radiometals and their chelators (siderophores), which are small molecules that bind radiometals and form a stable complex allowing sequestration by microbes. This radiometal-chelator complex can be directed to a specific microbial target in vivo, facilitating anatomical localization by PET or single photon emission computed tomography. Additionally, bifunctional chelators can further conjugate therapeutic molecules (e.g., peptides, antibiotics, antibodies) while still bound to desired radiometals, combining specific imaging with highly targeted antimicrobial therapy. These novel therapeutics may prove a useful complement to the armamentarium in the global fight against antimicrobial resistance. This review will highlight current state of infection imaging diagnostics and their limitations, strategies to develop infection-specific diagnostics, recent advances in radiometal-based chelators for microbial infection imaging, challenges, and future directions to improve targeted diagnostics and/or therapeutics.
Collapse
Affiliation(s)
- Asma Akter
- Department of Analytical, Environmental and Forensic Sciences, King’s College London, London, United Kingdom
| | - Oliver Lyons
- Vascular Endovascular and Transplant Surgery, Christchurch Public Hospital, Christchurch, New Zealand
- Department of Surgery, University of Otago, Christchurch, New Zealand
| | - Varun Mehra
- Department of Hematology, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Heather Isenman
- Department of Infectious Diseases, General Medicine, Christchurch Hospital, Christchurch, New Zealand
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
26
|
Jiang Y, Fang S, Feng J, Ruan Q, Zhang J. Synthesis and Bioevaluation of Novel Technetium-99m-Labeled Complexes with Norfloxacin HYNIC Derivatives for Bacterial Infection Imaging. Mol Pharm 2023; 20:630-640. [PMID: 36398935 DOI: 10.1021/acs.molpharmaceut.2c00830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To seek a novel 99mTc-labeled quinolone derivative for bacterial infection SPECT imaging that aims to lower nontarget organ uptake, a novel norfloxacin 6-hydrazinoicotinamide (HYNIC) derivative (HYNICNF) was designed and synthesized. It was radiolabeled with different coligands, such as tricine, trisodium triphenylphosphine-3,3',3″-trisulfonate (TPPTS), sodium triphenylphosphine-3-monosulfonate (TPPMS), and ethylenediamine-N,N'-diacetic acid (EDDA), to obtain three 99mTc-labeled norfloxacin HYNIC complexes, namely, [99mTc]Tc-tricine-TPPTS-HYNICNF, [99mTc]Tc-tricine-TPPMS-HYNICNF, and [99mTc]Tc-EDDA-HYNICNF. These complexes were purified (RCP > 95%) and evaluated in vitro and in vivo for targeting bacteria. All three complexes are hydrophilic, maintain good stability, and specifically bind Staphylococcus aureus in vitro. The biodistribution in mice with bacterial infection demonstrated that [99mTc]Tc-EDDA-HYNICNF showed a higher abscess uptake and lower nontarget organ uptake and was able to distinguish bacterial infection and sterile inflammation. Single photon emission computed tomography (SPECT) image study in bacterial infection mice showed there was a visible accumulation in the infection site, suggesting that [99mTc]Tc-EDDA-HYNICNF is a potential radiotracer for bacterial infection imaging.
Collapse
Affiliation(s)
- Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Si'an Fang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Product Administration), College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
27
|
Zhang Y, Hao M, Li L, Luo Q, Deng S, Yang Y, Liu Y, Fang W, Song E. Research progress of contrast agents for bacterial infection imaging in vivo. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
28
|
Gouws AC, Kruger HG, Gheysens O, Zeevaart JR, Govender T, Naicker T, Ebenhan T. Antibiotic-Derived Radiotracers for Positron Emission Tomography: Nuclear or "Unclear" Infection Imaging? Angew Chem Int Ed Engl 2022; 61:e202204955. [PMID: 35834311 PMCID: PMC9826354 DOI: 10.1002/anie.202204955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Indexed: 01/11/2023]
Abstract
The excellent features of non-invasive molecular imaging, its progressive technology (real-time, whole-body imaging and quantification), and global impact by a growing infrastructure for positron emission tomography (PET) scanners are encouraging prospects to investigate new concepts, which could transform clinical care of complex infectious diseases. Researchers are aiming towards the extension beyond the routinely available radiopharmaceuticals and are looking for more effective tools that interact directly with causative pathogens. We reviewed and critically evaluated (challenges or pitfalls) antibiotic-derived PET radiopharmaceutical development efforts aimed at infection imaging. We considered both radiotracer development for infection imaging and radio-antibiotic PET imaging supplementing other tools for pharmacologic drug characterization; overall, a total of 20 original PET radiotracers derived from eleven approved antibiotics.
Collapse
Affiliation(s)
- Arno Christiaan Gouws
- Catalysis and Peptide Research UnitUniversity of KwaZulu-NatalDurban4000South Africa
| | | | - Olivier Gheysens
- Department of Nuclear MedicineCliniques Universitaires Saint-Luc, and Institute of Clinical and Experimental ResearchUniversité Catholique de LouvainBrusselsBelgium
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure NPCPretoria0001South Africa
- RadiochemistryThe South African Nuclear Energy CorporationBrits0420South Africa
- Preclinical Drug Development PlatformNorth West UniversityPotchefstroom2520South Africa
| | | | - Tricia Naicker
- Catalysis and Peptide Research UnitUniversity of KwaZulu-NatalDurban4000South Africa
| | - Thomas Ebenhan
- Nuclear Medicine Research Infrastructure NPCPretoria0001South Africa
- Preclinical Drug Development PlatformNorth West UniversityPotchefstroom2520South Africa
- Department of Nuclear MedicineUniversity of PretoriaPretoria0001South Africa
| |
Collapse
|
29
|
Lindsley C, Müller CE, Bongarzone S. Diagnostic and Therapeutic Radiopharmaceuticals. ACS Pharmacol Transl Sci 2022; 5:835-837. [PMID: 36268118 PMCID: PMC9578137 DOI: 10.1021/acsptsci.2c00173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Craig
W. Lindsley
- Department
of Pharmacology, Department of Chemistry, and Vanderbilt Institute
of Chemical Biology, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Christa E. Müller
- PharmaCenter
Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität Bonn, D-53121 Bonn, Germany
| | - Salvatore Bongarzone
- Technical
Research and Development, Advanced Accelerator
Applications, a Novartis Company, via Ribes 5, Colleretto
Giacosa 10010, Italy
| |
Collapse
|
30
|
Lindsley CW, Müller CE, Bongarzone S. Diagnostic and Therapeutic Radiopharmaceuticals. J Med Chem 2022; 65:12497-12499. [DOI: 10.1021/acs.jmedchem.2c01403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Craig W. Lindsley
- Department of Pharmacology, Department of Chemistry, and Vanderbilt Institute of Chemical Biology, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität Bonn, D-53121 Bonn, Germany
| | - Salvatore Bongarzone
- Technical Research and Development, Advanced Accelerator Applications, a Novartis Company, via Ribes 5, Colleretto Giacosa 10010, Italy
| |
Collapse
|
31
|
Gouws AC, Kruger HG, Gheysens O, Zeevaart JR, Govender T, Naiker T, Ebenhan T. Antibiotic‐Derived Radiotracers for Positron Emission Tomography: Nuclear or ‘Unclear’ Infection Imaging? Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Arno Christiaan Gouws
- University of KwaZulu-Natal School of Health Sciences Catalysis and Peptide Research Unit SOUTH AFRICA
| | - Hendrik Gerhardus Kruger
- University of KwaZulu-Natal School of Health Sciences Catalysis and Peptide Research Unit SOUTH AFRICA
| | - Olivier Gheysens
- Cliniques Universitaires Saint-Luc Department of Nuclear Medicine BELGIUM
| | - Jan Rijn Zeevaart
- North-West University Potchefstroom Campus: North-West University Preclinical Drug Development Platform SOUTH AFRICA
| | | | - Tricia Naiker
- University of KwaZulu-Natal School of Health Sciences Catalysis and Peptide Research Unit SOUTH AFRICA
| | - Thomas Ebenhan
- University of Pretoria Nuclear Medicine Steve Biko and Malherbe St 0001 Pretoria SOUTH AFRICA
| |
Collapse
|
32
|
Peukert C, Popat Gholap S, Green O, Pinkert L, van den Heuvel J, van Ham M, Shabat D, Brönstrup M. Enzyme-Activated, Chemiluminescent Siderophore-Dioxetane Probes Enable the Selective and Highly Sensitive Detection of Bacterial Pathogens. Angew Chem Int Ed Engl 2022; 61:e202201423. [PMID: 35358362 PMCID: PMC9322335 DOI: 10.1002/anie.202201423] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Indexed: 12/18/2022]
Abstract
The sensitive detection of bacterial infections is a prerequisite for their successful treatment. The use of a chemiluminescent readout was so far hampered by an insufficient probe enrichment at the pathogens. We coupled siderophore moieties, that harness the unique iron transport system of bacteria, with enzyme-activatable dioxetanes and obtained seven trifunctional probes with high signal-to-background ratios (S/B=426-859). Conjugates with efficient iron transport capability into bacteria were identified through a growth recovery assay. All ESKAPE pathogens were labelled brightly by desferrioxamine conjugates, while catechols were weaker due to self-quenching. Bacteria could also be detected inside lung epithelial cells. The best probe 8 detected 9.1×103 CFU mL-1 of S. aureus and 5.0×104 CFU mL-1 of P. aeruginosa, while the analogous fluorescent probe 10 was 205-305fold less sensitive. This qualifies siderophore dioxetane probes for the selective and sensitive detection of bacteria.
Collapse
Affiliation(s)
- Carsten Peukert
- Department of Chemical BiologyHelmholtz Centre for Infection ResearchInhoffenstrasse 738124BraunschweigGermany
| | - Sachin Popat Gholap
- School of ChemistryRaymond and Beverly Sackler Faculty of Exact SciencesTel Aviv UniversityTel Aviv69978Israel
| | - Ori Green
- School of ChemistryRaymond and Beverly Sackler Faculty of Exact SciencesTel Aviv UniversityTel Aviv69978Israel
| | - Lukas Pinkert
- Department of Chemical BiologyHelmholtz Centre for Infection ResearchInhoffenstrasse 738124BraunschweigGermany
| | - Joop van den Heuvel
- Department of Structure and Function of ProteinsHelmholtz Centre for Infection ResearchInhoffenstrasse 738124BraunschweigGermany
| | - Marco van Ham
- Department of Structure and Function of ProteinsHelmholtz Centre for Infection ResearchInhoffenstrasse 738124BraunschweigGermany
| | - Doron Shabat
- School of ChemistryRaymond and Beverly Sackler Faculty of Exact SciencesTel Aviv UniversityTel Aviv69978Israel
| | - Mark Brönstrup
- Department of Chemical BiologyHelmholtz Centre for Infection ResearchInhoffenstrasse 738124BraunschweigGermany
| |
Collapse
|
33
|
Erba PA, Bartoli F, Sollini M, Raffaella B, Zanca R, Enrica E, Lazzeri E. Alternative Nuclear Imaging Tools for Infection Imaging. Curr Cardiol Rep 2022; 24:879-891. [PMID: 35696046 PMCID: PMC9288362 DOI: 10.1007/s11886-022-01708-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 11/30/2022]
Abstract
Purpose of Review Cardiovascular infections are serious disease associated with high morbidity and mortality. Their diagnosis is challenging, requiring a proper management for a prompt recognition of the clinical manifestations, and a multidisciplinary approach involving cardiologists, cardiothoracic surgeons, infectious diseases specialist, imagers, and microbiologists. Imaging plays a central role in the diagnostic workout, including molecular imaging techniques. In this setting, two different strategies might be used to image infections: the first is based on the use of agents targeting the microorganism responsible for the infection. Alternatively, we can target the components of the pathophysiological changes of the inflammatory process and/or the host response to the infectious pathogen can be considered. Understanding the strength and limitations of each strategy is crucial to select the most appropriate imaging tool. Recent Findings Currently, multislice computed tomography (MSCT) and nuclear imaging (18F-fluorodeoxyglucose positron emission tomography/computed tomography, and leucocyte scintigraphy) are part of the diagnostic strategies. The main role of nuclear medicine imaging (PET/CT and SPECT/CT) is the confirmation of valve/CIED involvement and/or associated perivalvular infection and the detection of distant septic embolism. Proper patients’ preparation, imaging acquisition, and reconstruction as well as imaging reading are crucial to maximize the diagnostic information. Summary In this manuscript, we described the use of molecular imaging techniques, in particular WBC imaging, in patients with infective endocarditis, cardiovascular implantable electronic device infections, and infections of composite aortic graft, underlying the strength and limitations of such approached as compared to the other imaging modalities.
Collapse
Affiliation(s)
- Paola Anna Erba
- Regional Center of Nuclear Medicine, Department of Translational Research and Advanced Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, 56126, Pisa, Italy.
- Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Francesco Bartoli
- Regional Center of Nuclear Medicine, Department of Translational Research and Advanced Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Berchiolli Raffaella
- Vascular Surgery Unit, Department of Translational Research and Advanced Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Roberta Zanca
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Esposito Enrica
- Regional Center of Nuclear Medicine, Department of Translational Research and Advanced Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - Elena Lazzeri
- Regional Center of Nuclear Medicine, Department of Translational Research and Advanced Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, 56126, Pisa, Italy
| |
Collapse
|
34
|
Firth G, Blower JE, Bartnicka JJ, Mishra A, Michaels AM, Rigby A, Darwesh A, Al-Salemee F, Blower PJ. Non-invasive radionuclide imaging of trace metal trafficking in health and disease: "PET metallomics". RSC Chem Biol 2022; 3:495-518. [PMID: 35656481 PMCID: PMC9092424 DOI: 10.1039/d2cb00033d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/07/2022] [Indexed: 12/05/2022] Open
Abstract
Several specific metallic elements must be present in the human body to maintain health and function. Maintaining the correct quantity (from trace to bulk) and location at the cell and tissue level is essential. The study of the biological role of metals has become known as metallomics. While quantities of metals in cells and tissues can be readily measured in biopsy and autopsy samples by destructive analytical techniques, their trafficking and its role in health and disease are poorly understood. Molecular imaging with radionuclides - positron emission tomography (PET) and single photon emission computed tomography (SPECT) - is emerging as a means to non-invasively study the acute trafficking of essential metals between organs, non-invasively and in real time, in health and disease. PET scanners are increasingly widely available in hospitals, and methods for producing radionuclides of some of the key essential metals are developing fast. This review summarises recent developments in radionuclide imaging technology that permit such investigations, describes the radiological and physicochemical properties of key radioisotopes of essential trace metals and useful analogues, and introduces current and potential future applications in preclinical and clinical investigations to study the biology of essential trace metals in health and disease.
Collapse
Affiliation(s)
- George Firth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Julia E Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Joanna J Bartnicka
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Aishwarya Mishra
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Aidan M Michaels
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Alex Rigby
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Afnan Darwesh
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Fahad Al-Salemee
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| |
Collapse
|
35
|
Peukert C, Gholap SP, Green O, Pinkert L, van den Heuvel J, van Ham M, Shabat D, Broenstrup M. Enzyme‐activated, Chemiluminescent Siderophore‐Dioxetane Probes Enable the Selective and Highly Sensitive Detection of Bacterial ESKAPE Pathogens. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202201423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Carsten Peukert
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Chemical Biology GERMANY
| | - Sachin Popat Gholap
- Tel Aviv University Raymond and Beverly Sackler Faculty of Exact Sciences School of Chemistry ISRAEL
| | - Ori Green
- Tel Aviv University Raymond and Beverly Sackler Faculty of Exact Sciences School of Chemistry ISRAEL
| | - Lukas Pinkert
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH Chemical Biology GERMANY
| | - Joop van den Heuvel
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH SFPR GERMANY
| | - Marco van Ham
- Helmholtz-Zentrum für Infektionsforschung GmbH: Helmholtz-Zentrum fur Infektionsforschung GmbH SFPR GERMANY
| | - Doron Shabat
- Tel Aviv University Raymond and Beverly Sackler Faculty of Exact Sciences School of Chemistry ISRAEL
| | - Mark Broenstrup
- Helmholtz-Zentrum fur Infektionsforschung GmbH Chemical Biology Inhoffenstraße 7 38124 Braunschweig GERMANY
| |
Collapse
|
36
|
Muranaka Y, Mizutani A, Kobayashi M, Nakamoto K, Matsue M, Nishi K, Yamazaki K, Nishii R, Shikano N, Okamoto S, Kawai K. Comparison of L- and D-Amino Acids for Bacterial Imaging in Lung Infection Mouse Model. Int J Mol Sci 2022; 23:ijms23052467. [PMID: 35269610 PMCID: PMC8910731 DOI: 10.3390/ijms23052467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The effectiveness of L- and D-amino acids for detecting the early stage of infection in bacterial imaging was compared. We evaluated the accumulation of 3H-L-methionine (Met), 3H-D-Met, 3H-L-alanine (Ala), and 3H-D-Ala in E. coli EC-14 and HaCaT cells. Biological distribution was assessed in control and lung-infection-model mice with EC-14 using 3H-L- and D-Met, and 18F-FDG. A maximum accumulation of 3H-L- and D-Met, and 3H-L- and D-Ala occurred in the growth phase of EC-14 in vitro. The accumulation of 3H-L-Met and L-Ala was greater than that of 3H-D-Met and D-Ala in both EC-14 and HaCaT cells. For all radiotracers, the accumulation was greater in EC-14 than in HaCaT cells at early time points. The accumulation was identified at 5 min after injection in EC-14, whereas the accumulation gradually increased in HaCaT cells over time. There was little difference in biodistribution between 3H-L-and D-Met except in the brain. 3H-L- and D-Met were sensitive for detecting areas of infection after the spread of bacteria throughout the body, whereas 18F-FDG mainly detected primary infection areas. Therefore, 11C-L- and D-Met, radioisotopes that differ only in terms of 3H labeling, could be superior to 18F-FDG for detecting bacterial infection in lung-infection-model mice.
Collapse
Affiliation(s)
- Yuka Muranaka
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Ishikawa, Japan; (Y.M.); (K.N.)
| | - Asuka Mizutani
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Ishikawa, Japan; (A.M.); (M.K.); (S.O.)
| | - Masato Kobayashi
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Ishikawa, Japan; (A.M.); (M.K.); (S.O.)
| | - Koya Nakamoto
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Ishikawa, Japan; (Y.M.); (K.N.)
| | - Miki Matsue
- Ishikawa Prefectural Institute of Public Health and Environmental Science, 1-11, Taiyogaoka, Kanazawa 920-1154, Ishikawa, Japan;
| | - Kodai Nishi
- Department of Radioisotope Medicine, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Nagasaki, Japan;
| | - Kana Yamazaki
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage, Chiba 263-8555, Chiba, Japan; (K.Y.); (R.N.)
| | - Ryuichi Nishii
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage, Chiba 263-8555, Chiba, Japan; (K.Y.); (R.N.)
| | - Naoto Shikano
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences, 4669-2 Ami, Inashiki 300-0394, Ibaraki, Japan;
| | - Shigefumi Okamoto
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Ishikawa, Japan; (A.M.); (M.K.); (S.O.)
- Advanced Health Care Science Research Unit, Innovative Integrated Bio-Research Core Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-0942, Ishikawa, Japan
| | - Keiichi Kawai
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Ishikawa, Japan; (A.M.); (M.K.); (S.O.)
- Biomedical Imaging Research Center, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji 910-1193, Fukui, Japan
- Correspondence: ; Tel.: +81-76-265-2527; Fax: +81-76-234-4366
| |
Collapse
|
37
|
MicroPET imaging of bacterial infection with nitroreductase-specific responsive 18F-labelled nitrogen mustard analogues. Eur J Nucl Med Mol Imaging 2022; 49:2645-2654. [PMID: 35122512 DOI: 10.1007/s00259-022-05710-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/26/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Bacterial infection and antibiotic resistance are serious threats to human health. This study aimed to develop two novel radiotracers, 18F-NTRP and 18F-NCRP, that possess a specific nitroreductase (NTR) response to image deep-seated bacterial infections using positron emission tomography (PET). This method can distinguish infection from sterile inflammation. METHODS 18F-NTRP and 18F-NCRP were synthesized via a one-step method; all the steps usually involved in tracer radiosynthesis were successfully adapted in the All-In-One automated module. After the physiochemical properties of 18F-NTRP and 18F-NCRP were characterized, their specificity and selectivity for NTR were verified in E. coli and S. aureus. The ex vivo biodistribution of the tracers was evaluated in normal mice. MicroPET-CT imaging was performed in mouse models of bacterial infection and inflammation after the administration of 18F-NTRP or 18F-NCRP. RESULTS Fully automated radiosynthesis of 18F-NTRP and 18F-NCRP was achieved within 90-110 min with overall decay-uncorrected, isolated radiochemical yields of 21.24 ± 4.25% and 11.3 ± 3.78%, respectively. The molar activities of 18F-NTRP and 18F-NCRP were 320 ± 40 GBq/μmol and 275 ± 33 GBq/µmol, respectively. In addition, 18F-NTRP and 18F-NCRP exhibited high selectivity and specificity for NTR response. PET-CT imaging in bacteria-infected mouse models with 18F-NTRP or 18F-NCRP showed significant radioactivity uptake in either E. coli- or S. aureus-infected muscles. The uptake for E. coli-infected muscles, 2.4 ± 0.2%ID/g with 18F-NTRP and 4.05 ± 0.49%ID/g with 18F-NCRP, was up to three times greater than that for uninfected control muscles. Furthermore, for both 18F-NTRP and 18F-NCRP, the uptake in bacterial infection was 2.6 times higher than that in sterile inflammation, allowing an effective distinction of infection from inflammation. CONCLUSION 18F-NTRP and 18F-NCRP are worth further investigation to verify their potential clinical application for distinguishing bacterial infection from sterile inflammation via their specific NTR responsiveness.
Collapse
|
38
|
Ruiz-Bedoya CA, Mota F, Ordonez AA, Foss CA, Singh AK, Praharaj M, Mahmud FJ, Ghayoor A, Flavahan K, De Jesus P, Bahr M, Dhakal S, Zhou R, Solis CV, Mulka KR, Bishai WR, Pekosz A, Mankowski JL, Villano J, Klein SL, Jain SK. 124I-Iodo-DPA-713 Positron Emission Tomography in a Hamster Model of SARS-CoV-2 Infection. Mol Imaging Biol 2022; 24:135-143. [PMID: 34424479 PMCID: PMC8381721 DOI: 10.1007/s11307-021-01638-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Molecular imaging has provided unparalleled opportunities to monitor disease processes, although tools for evaluating infection remain limited. Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is mediated by lung injury that we sought to model. Activated macrophages/phagocytes have an important role in lung injury, which is responsible for subsequent respiratory failure and death. We performed pulmonary PET/CT with 124I-iodo-DPA-713, a low-molecular-weight pyrazolopyrimidine ligand selectively trapped by activated macrophages cells, to evaluate the local immune response in a hamster model of SARS-CoV-2 infection. PROCEDURES Pulmonary 124I-iodo-DPA-713 PET/CT was performed in SARS-CoV-2-infected golden Syrian hamsters. CT images were quantified using a custom-built lung segmentation tool. Studies with DPA-713-IRDye680LT and a fluorescent analog of DPA-713 as well as histopathology and flow cytometry were performed on post-mortem tissues. RESULTS Infected hamsters were imaged at the peak of inflammatory lung disease (7 days post-infection). Quantitative CT analysis was successful for all scans and demonstrated worse pulmonary disease in male versus female animals (P < 0.01). Increased 124I-iodo-DPA-713 PET activity co-localized with the pneumonic lesions. Additionally, higher pulmonary 124I-iodo-DPA-713 PET activity was noted in male versus female hamsters (P = 0.02). DPA-713-IRDye680LT also localized to the pneumonic lesions. Flow cytometry demonstrated a higher percentage of myeloid and CD11b + cells (macrophages, phagocytes) in male versus female lung tissues (P = 0.02). CONCLUSION 124I-Iodo-DPA-713 accumulates within pneumonic lesions in a hamster model of SARS-CoV-2 infection. As a novel molecular imaging tool, 124I-Iodo-DPA-713 PET could serve as a noninvasive, clinically translatable approach to monitor SARS-CoV-2-associated pulmonary inflammation and expedite the development of novel therapeutics for COVID-19.
Collapse
Affiliation(s)
- Camilo A Ruiz-Bedoya
- 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alvaro A Ordonez
- 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Catherine A Foss
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alok K Singh
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Monali Praharaj
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Farina J Mahmud
- 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Kelly Flavahan
- 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Melissa Bahr
- 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Clarisse V Solis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen R Mulka
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William R Bishai
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jason Villano
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, 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, 1550 Orleans Street, CRB-II Room 109, Baltimore, MD, USA.
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
39
|
Ordonez AA, Parker MF, Miller RJ, Plyku D, Ruiz-Bedoya CA, Tucker EW, Luu JM, Dikeman DA, Lesniak WG, Holt DP, Dannals RF, Miller LS, Rowe SP, Wilson DM, Jain SK. 11C-Para-aminobenzoic acid PET imaging of S. aureus and MRSA infection in preclinical models and humans. JCI Insight 2022; 7:154117. [PMID: 35014627 PMCID: PMC8765043 DOI: 10.1172/jci.insight.154117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Tools for noninvasive detection of bacterial pathogens are needed but are not currently available for clinical use. We have previously shown that para-aminobenzoic acid (PABA) rapidly accumulates in a wide range of pathogenic bacteria, motivating the development of related PET radiotracers. In this study, 11C-PABA PET imaging was used to accurately detect and monitor infections due to pyogenic bacteria in multiple clinically relevant animal models. 11C-PABA PET imaging selectively detected infections in muscle, intervertebral discs, and methicillin-resistant Staphylococcus aureus–infected orthopedic implants. In what we believe to be first-in-human studies in healthy participants, 11C-PABA was safe, well-tolerated, and had a favorable biodistribution, with low background activity in the lungs, muscles, and brain. 11C-PABA has the potential for clinical translation to detect and localize a broad range of bacteria.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research and.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew Fl Parker
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | | | - Donika Plyku
- Russell H. Morgan Department of Radiology and Radiological Sciences, and
| | - Camilo A Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research and.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth W Tucker
- Center for Infection and Inflammation Imaging Research and.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Justin M Luu
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | | | - Wojciech G Lesniak
- Russell H. Morgan Department of Radiology and Radiological Sciences, and
| | - Daniel P Holt
- Russell H. Morgan Department of Radiology and Radiological Sciences, and
| | - Robert F Dannals
- Russell H. Morgan Department of Radiology and Radiological Sciences, and
| | | | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Sciences, and
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research and.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Russell H. Morgan Department of Radiology and Radiological Sciences, and
| |
Collapse
|
40
|
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: 16] [Impact Index Per Article: 4.0] [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
|
41
|
Li L, Gu P, Hao M, Xiang X, Feng Y, Zhu X, Song Y, Song E. Bacteria-Targeted MRI Probe-Based Imaging Bacterial Infection and Monitoring Antimicrobial Therapy In Vivo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103627. [PMID: 34554653 DOI: 10.1002/smll.202103627] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/24/2021] [Indexed: 06/13/2023]
Abstract
Despite the significant advances of imaging techniques nowadays, accurate diagnosis of bacterial infections and real-time monitoring the efficacy of antibiotic therapy in vivo still remain huge challenges. Herein, a self-assembling peptide (FFYEGK) and vancomycin (Van) antibiotic molecule co-modified gadolinium (Gd) MRI nanoaggregate probe (GFV) for detecting Staphylococcus aureus (S. aureus) infection in vivo and monitoring the treatment of S. aureus-infected myositis by using daptomycin (Dap) antibiotic as model are designed and fabricated. The as-prepared GFV probe bears Van molecules, making itself good bacteria-specific targeting, and the peptide in the probe can enhance the longitudinal relaxivity rate (r1 ) after self-assembly due to the π-π stacking. The study showed that, based on the GFV probe, bacterial infections and sterile inflammation can be discriminated, and as few as 105 cfu S. aureus can be detected in vivo with high specificity and accurately. Moreover, the T1 signal of GFV probe at the S. aureus-infected site in mice correlates with the increasing time of Dap treating, indicating the possibility of monitoring the efficacy of antibacterial agents for infected mice based on the as proposed GFV probe. This study shows the potential of GFV probe for diagnosis, evaluation, and prognosis of infectious diseases in clinics.
Collapse
Affiliation(s)
- Linyao Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Peilin Gu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Mengqi Hao
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Xiaoli Xiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Yuting Feng
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Xiaokang Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, CAS, Beijing, 100085, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
42
|
Ataeinia B, Heidari P. Artificial Intelligence and the Future of Diagnostic and Therapeutic Radiopharmaceutical Development:: In Silico Smart Molecular Design. PET Clin 2021; 16:513-523. [PMID: 34364818 PMCID: PMC8453048 DOI: 10.1016/j.cpet.2021.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Novel diagnostic and therapeutic radiopharmaceuticals are increasingly becoming a central part of personalized medicine. Continued innovation in the development of new radiopharmaceuticals is key to sustained growth and advancement of precision medicine. Artificial intelligence has been used in multiple fields of medicine to develop and validate better tools for patient diagnosis and therapy, including in radiopharmaceutical design. In this review, we first discuss common in silico approaches and focus on their usefulness and challenges in radiopharmaceutical development. Next, we discuss the practical applications of in silico modeling in design of radiopharmaceuticals in various diseases.
Collapse
Affiliation(s)
- Bahar Ataeinia
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA
| | - Pedram Heidari
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA.
| |
Collapse
|
43
|
Peukert C, Langer LNB, Wegener SM, Tutov A, Bankstahl JP, Karge B, Bengel FM, Ross TL, Brönstrup M. Optimization of Artificial Siderophores as 68Ga-Complexed PET Tracers for In Vivo Imaging of Bacterial Infections. J Med Chem 2021; 64:12359-12378. [PMID: 34370949 DOI: 10.1021/acs.jmedchem.1c01054] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The diagnosis of bacterial infections at deep body sites benefits from noninvasive imaging of molecular probes that can be traced by positron emission tomography (PET). We specifically labeled bacteria by targeting their iron transport system with artificial siderophores. The cyclen-based probes contain different binding sites for iron and the PET nuclide gallium-68. A panel of 11 siderophores with different iron coordination numbers and geometries was synthesized in up to 8 steps, and candidates with the best siderophore potential were selected by a growth recovery assay. The probes [68Ga]7 and [68Ga]15 were found to be suitable for PET imaging based on their radiochemical yield, radiochemical purity, and complex stability in vitro and in vivo. Both showed significant uptake in mice infected with Escherichia coli and were able to discern infection from lipopolysaccharide-triggered, sterile inflammation. The study qualifies cyclen-based artificial siderophores as readily accessible scaffolds for the in vivo imaging of bacteria.
Collapse
Affiliation(s)
- Carsten Peukert
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Laura N B Langer
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Sophie M Wegener
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anna Tutov
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Bianka Karge
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Center for Biomolecular Drug Research (BMWZ), Schneiderberg 38, 30167 Hannover, Germany
| |
Collapse
|
44
|
Naqvi SAR. 99m Tc-labeled antibiotics for infection diagnosis: Mechanism, action, and progress. Chem Biol Drug Des 2021; 99:56-74. [PMID: 34265177 DOI: 10.1111/cbdd.13923] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/08/2021] [Accepted: 06/06/2021] [Indexed: 01/01/2023]
Abstract
Discovery of penicillin marked a turning point in the history of infection therapy which also led to the emergence of bacterial resistance. It is now 100 years to fight with ever-muted variants of pathogens by developing more and more antibiotics. Since 1987 to todate, no successful class of antibiotic was introduced; this three decade period is known as "the discovery void" period. While, the clinically approved antibiotics are gradually dying in front of bacterial resistance due to which bacterial infections are appearing leading cause of death and disability. Nuclear medicine imaging technique is the strongest modality to diagnose and follow-up of deep-seated and complicated infections. However, the selection of radiolabeled antimicrobial agents plays critical role in gaining sensitivity and specificity of the imaging results. This review comprises of two main sections; first section explains antibiotic targets, and second section explains the imaging efficacy of 99m Tc-labeled antimicrobial agents against bacterial infection along with the emphasis on progress and update of 99m Tc-labeled antibiotics as infection imaging probes. The review, in conclusion, could be an acceleration for radiopharmaceutical chemists for designing and developing 99m Tc-labeled antimicrobial agents to improve infection imaging quality.
Collapse
Affiliation(s)
- Syed Ali Raza Naqvi
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
45
|
Wang X, Wang Q, Zhang Q, Han X, Xu S, Yin D, Hu HY. Developing fluoromodule-based probes for in vivo monitoring the bacterial infections and antibiotic responses. Talanta 2021; 233:122610. [PMID: 34215094 DOI: 10.1016/j.talanta.2021.122610] [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: 04/02/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
Recently, antibiotic resistant has become a serious public health concern, which warrants new generations of antibiotics to be developed. Pharmacodynamic evaluation is crucial in drug discovery processes. Despite numerous advanced imaging systems are available nowadays, technologies for the sensitive in vivo diagnosis of bacterial infections and direct visualization of drug efficacy are yet to be developed. In this study, we have developed novel near-infrared (NIR) fluorogenic probes. These probes are dark in solution but highly fluorescent when bound to the cognate reporter, fluorogen-activating protein (FAP). We established the in vivo bacterial infection model using FAP_dH6.2 recombinantly expressed E. coli and applied this NIR fluoromodule-based system for diagnosing bacterial infections and monitoring disease progressions and its responses to a type of antibiotics through classic mechanism of membrane lysis. This NIR fluoromodule-based system will discover new information on bacterial infections and identify newer antibacterial entities.
Collapse
Affiliation(s)
- Xiang Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Qinghua Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Qingyang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiaowan Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Shengnan Xu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
46
|
Synthesis and Evaluation of Novel Norfloxacin Isonitrile 99mTc Complexes as Potential Bacterial Infection Imaging Agents. Pharmaceutics 2021; 13:pharmaceutics13040518. [PMID: 33918583 PMCID: PMC8069222 DOI: 10.3390/pharmaceutics13040518] [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: 03/16/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 11/17/2022] Open
Abstract
To develop potential technetium-99m single-photon emission computed tomography (SPECT) imaging agents for bacterial infection imaging, the novel norfloxacin isonitrile derivatives CN4NF and CN5NF were synthesized and radiolabeled with a [99mTc][Tc(I)]+ core to obtain [99mTc]Tc-CN4NF and [99mTc]Tc-CN5NF. These compounds were produced in high radiolabeling yields and showed hydrophilicity and good stability in vitro. The bacterial binding assay indicated that [99mTc]Tc-CN4NF and [99mTc]Tc-CN5NF were specific to bacteria. Compared with [99mTc]Tc-CN4NF, biodistribution studies of [99mTc]Tc-CN5NF showed a higher uptake in bacteria-infected tissues than in turpentine-induced abscesses, indicating that [99mTc]Tc-CN5NF could distinguish bacterial infection from sterile inflammation. In addition, [99mTc]Tc-CN5NF had higher abscess/blood and abscess/muscle ratios. SPECT image of [99mTc]Tc-CN5NF showed that there was a clear accumulation in the infection site, suggesting that it could be a potential bacterial infection imaging radiotracer.
Collapse
|
47
|
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: 1.5] [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
|
48
|
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
|
49
|
Fang S, Jiang Y, Gan Q, Ruan Q, Xiao D, Zhang J. Design, Preparation, and Evaluation of a Novel 99mTcN Complex of Ciprofloxacin Xanthate as a Potential Bacterial Infection Imaging Agent. Molecules 2020; 25:molecules25245837. [PMID: 33322004 PMCID: PMC7762968 DOI: 10.3390/molecules25245837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 01/17/2023] Open
Abstract
In order to seek novel technetium-99m bacterial infection imaging agents, a ciprofloxacin xanthate (CPF2XT) was synthesized and radiolabeled with [99mTcN]2+ core to obtain the 99mTcN-CPF2XT complex, which exhibited high radiochemical purity, hydrophilicity, and good stability in vitro. The bacteria binding assay indicated that 99mTcN-CPF2XT had specificity to bacteria. A study of biodistribution in mice showed that 99mTcN-CPF2XT had a higher uptake in bacterial infection tissues than in turpentine-induced abscesses, indicating that it could distinguish bacterial infection from sterile inflammation. Compared to 99mTcN-CPFXDTC, the abscess/blood and abscess/muscle ratios of 99mTcN-CPF2XT were higher and the uptakes of 99mTcN-CPF2XT in the liver and lung were obviously decreased. The results suggested that 99mTcN-CPF2XT would be a potential bacterial infection imaging agent.
Collapse
|
50
|
Saccharomyces cerevisiae and Candida albicans Yeast Cells Labeled with Fe(III) Complexes as MRI Probes. MAGNETOCHEMISTRY 2020. [DOI: 10.3390/magnetochemistry6030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The development of MRI probes is of interest for labeling antibiotic-resistant fungal infections based on yeast. Our work showed that yeast cells can be labeled with high-spin Fe(III) complexes to produce enhanced T2 water proton relaxation. These Fe(III)-based macrocyclic complexes contained a 1,4,7-triazacyclononane framework, two pendant alcohol groups, and either a non-coordinating ancillary group and a bound water molecule or a third coordinating pendant. The Fe(III) complexes that had an open coordination site associated strongly with Saccharomyces cerevisiae upon incubation, as shown by screening using Z-spectra analysis. The incubation of one Fe(III) complex with either Saccharomyces cerevisiae or Candida albicans yeast led to an interaction with the β-glucan-based cell wall, as shown by the ready retrieval of the complex by the bidentate chelator called maltol. Other conditions, such as a heat shock treatment of the complexes, produced Fe(III) complex uptake that could not be reversed by the addition of maltol. Appending a fluorescence dye to Fe(TOB) led to uptake through secretory pathways, as shown by confocal fluorescence microscopy and by the incomplete retrieval of the Fe(III) complex by the maltol treatment. Yeast cells that were labeled with these Fe(III) complexes displayed enhanced water proton T2 relaxation, both for S. cerevisiae and for yeast and hyphal forms of C. albicans.
Collapse
|