Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Mar 1.
Published in final edited form as: Semin Nucl Med. 2017 Dec 14;48(2):182–194. doi: 10.1053/j.semnuclmed.2017.11.003

Pathogen-specific Bacterial Imaging in Nuclear Medicine

Alvaro A Ordonez 1,2, Sanjay K Jain 1,2
PMCID: PMC5819618  NIHMSID: NIHMS922675  PMID: 29452620

Abstract

When serious infections are suspected, patients are often treated empirically with broad-spectrum antibiotics while awaiting results that provide information on the bacterial class and species causing the infection, as well as drug susceptibilities. For deep-seated infections, these traditional diagnostic techniques often rely on tissue biopsies to obtain clinical samples which can be expensive, dangerous and has the potential of sampling bias. Moreover, these procedures and results can take several days and may not always provide reliable information. This combination of time and effort required for proper antibiotic selection has become a barrier leading to indiscriminate broad-spectrum antibiotic use. Exposure to nosocomial infections and indiscriminate use of broad-spectrum antibiotics are responsible for promoting bacterial drug-resistance leading to substantial morbidity and mortality, especially in hospitalized and immunosuppressed patients. Therefore, early diagnosis of infection and targeted antibiotic treatments are urgently needed to reduce morbidity and mortality caused by bacterial infections worldwide. Reliable pathogen-specific bacterial imaging techniques have the potential to provide early diagnosis and guide antibiotic treatments.

INTRODUCTION

Bacterial infections are a major cause of morbidity and mortality worldwide. However, with antimicrobial resistance emerging and spreading globally, multi-drug resistant organisms (MDRO) are an increasingly serious threat to human health. In the United States alone, at least 2 million people become infected with MDRO bacteria each year, with at least 23,000 of these patients dying as a direct result of these infections, leading to significant social and economic costs.13 Although antimicrobial resistance can occur naturally, the misuse and overuse of antibiotics is accelerating this process.48 Improved diagnostic accuracy of pathogen-specific imaging techniques could curb the inappropriate use of antibiotics for non-infectious entities, streamline empiric antibiotic selection to target the class of the bacteria causing the infection and provide a way to rapidly monitor antibiotic treatments.

A common clinical problem for practicing infectious disease specialists and radiologists is differentiating active infection from other causes of inflammation. Current practice in microbiology (microbiologic cultures or molecular techniques) relies on the availability of a clinical sample (blood, urine, etc.). However, when a deep-seated infection is suspected, a biopsy is often required, leading to costly, invasive and sometimes dangerous procedures, prone to sampling errors. Therefore, clinicians frequently use conventional radiological techniques such as computerized tomography (CT), magnetic resonance imaging (MRI), X-ray, and ultrasound, to aid in the diagnosis and localization of deep-seated infections.9 However, these techniques rely on the presence of structural abnormalities in tissues and organs caused by the infection and/or inflammatory response of the host and are unable to reliably differentiate infection from inflammation or oncologic processes. To try to address this clinical challenge, multiple molecular imaging techniques have been developed to detect bacteria in vitro, in animal models, and for clinical studies. Extensive research has been done using optical imaging techniques with promising results,1014 and although these studies have helped significantly in understanding disease pathogenesis and in developing therapeutics for infections, their clinical evaluation for bacterial infections has been limited by tissue penetration. Conversely, nuclear imaging techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are not limited by tissue depth and use very small amounts of radioactive tracers that reflect the physiological and biochemical changes associated with the infection process.

Nuclear imaging of bacterial infections has been a developing field for more than 50 years and provides valuable insight not only in the diagnosis and monitoring of infections, but it can also help understand the disease pathogenesis in both preclinical and clinical settings.15 Although most of the published literature in this area involves non-specific imaging agents such as 18F-fluorodeoxyglucose (18F-FDG), 67Ga-citrate and in vitro radiolabeled leukocytes, significant efforts have been made to develop bacteria-specific radiolabeled tracers that can bind or accumulate only in bacteria.16 This is a challenging task since the ideal bacteria-specific imaging compound should be both sensitive and specific for detection of the infection site, in addition to being nontoxic, affordable, widely available, and easily and rapidly prepared (Table 1). These compounds should have ideal biochemical properties (e.g. moderate lipophilicity, low plasma protein binding, and metabolic stability),17,18 as well as optimal pharmacokinetic and pharmacodynamic features for maximum performance and usability.19 The tracer should be able to rapidly localize to the infection site with little non-specific binding and have an optimal clearance rate that allows sufficient time for it to reach its target but is rapid enough to excrete unbound probe and minimize radiation exposure.20 The search for the best bacteria-specific imaging tracers is still an ongoing challenge and this review summarizes the efforts made to achieve this goal.

Table 1.

Desirable properties of an ideal bacteria-specific PET imaging tracer (adapted from Gemmel et al.19 and Johnson et al.18)

Sensitive High target to background signal ratio. Low limit of detection.
Specific Probe targets infectious, not inflammatory lesions.
Quantitative Signal proportionate to infectious burden.
Rapid Fast localization to the site of infection.
Stable Probe retained at the site of infection. No degradation of the probe by the host.
Safe Acceptable radiation dose. Repeat injection feasible, without pharmacologic or immunologic effect.
Manufacturable Uncomplicated synthesis with reasonable expense.

METABOLISM-BASED TRACERS

Based on the principles utilized in clinical microbiology, which uses selective metabolism to differentiate microbes,21 bacteria-specific imaging tracers based on selective metabolic pathways expressed only in bacteria (or a specific class of bacteria) could be developed not only to detect the presence of bacterial infection, but also to identify the class of bacteria causing the infections.22 Metabolism-based tracers, such as radio-fluorinated sugars and sugar alcohols, combined with favorable radiation dosimetry due to short half-life isotopes (e.g. 18F), provide multiple advantages such as easy penetration into diseased tissues, possible accumulation inside bacteria and rapid clearance from non-target tissues. However, since these tracers are designed to only be incorporated into bacteria, there would be limitations on their use for diagnosis of pathogens with a predominantly intracellular lifecycle (e.g. Listeria monocytogenes). Similarly, the incorporation rate of these tracers into the cells could be affected by the metabolic state of the bacteria.

Nucleoside analogs

The thymidine analog FIAU (1-(2′-deoxy-2′-fluoro-β-D-arabinofuranosyl)-5-iodouracil) was initially developed as a non-invasive agent to non-invasively monitor tumor-targeting bacteria. FIAU is a substrate for the native thymidine kinases (TK) of herpes viruses and a wide variety of bacterial species. Once inside the bacterial cell, FIAU gets phosphorylated by TK and subsequently trapped. However, bacteria that do not possess the TK enzyme (e.g. Pseudomonas aeruginosa), would not retain FIAU.23 In 2004, Bettegowda et al. reported successful in vivo imaging of bacterial infections in mice using 125I-FIAU.24 Using a lung infection murine model, Pullambhatla et al. also showed that this tracer can be used to monitor the efficacy of antimicrobial therapy.25 Subsequent studies with 124I-FIAU reported promising results in patients with musculoskeletal infections (Fig. 1).26 However, a recent study found that 124I-FIAU lacks specificity in patients with prosthetic joint infections, with high background signal in uninfected muscle presumably due to host mitochondrial metabolism.27 Although the synthesis methods and dosimetry of 18F-FIAU in animal models have also been reported, data for bacterial imaging is not available.28

Figure 1. 124I-FIAU signal in established infections as imaged by PET/CT.

Figure 1

Fused PET and CT images, taken at 2 hours after radiotracer administration, are shown for the following cases: (a) septic arthritis (right knee), (b) septic arthritis (right knee), (c) osteomyelitis (left distal tibia), (d) cellulitis (left lower extremity), (e) necrotizing septic arthritis (left knee). Reprinted with permission by the Public Library of Science from Diaz et al. 2007.26

Sugars

While widely employed in clinical oncology, 18F-FDG is presently the most used PET tracer for imaging bacterial infections by identifying infection-associated inflammatory responses.29,30 Immune cells increase the use of glucose as an energy source during metabolic bursts associated with inflammatory responses due to infection.31 Even during chronic inflammation, increased cell glycolysis persists,32 giving 18F-FDG the potential to image infections with high sensitivity.31 Current guidelines recommend 18F-FDG PET imaging in clinical cases of peripheral bone osteomyelitis, suspected spinal infection and fever of unknown origin (FUO),33 and suggest it can be used in other infection settings. 18F-FDG PET has also been used to monitor tuberculosis (TB) patients,34,35 evaluate patients with inconclusive diagnosis of infective endocarditis,36 help distinguish between septic and aseptic loosening in joint prosthesis infection,37 and diagnose diabetic foot infections.38 However, since 18F-FDG is metabolized by the same pathways as glucose, and given the widespread expression of glucose surface transporters in eukaryotic cells, 18F-FDG is not able to differentiate true infection from sterile inflammation. Mills et al. developed the phosphate analog 18F-FDG-6-P in an effort to facilitate specific uptake of 18F-FDG by bacterial hexose phosphate transporters.39 However, in vivo PET imaging with 18F-FDG-6-P in a foreign body Staphylococcus aureus infection mouse model demonstrated that the biodistribution was similar to that of 18F-FDG, therefore limiting its use for bacteria-specific imaging.

Sorbitol, a sugar alcohol, is a metabolic substrate for Enterobacteriaceae (Klebsiella pneumoniae, Escherichia coli, Yersinia spp., Enterobacter spp., etc.) which are the largest group of Gram-negative bacterial pathogens in humans. Similar to glucose, sorbitol is selectively taken up by bacteria via surface transporters, phosphorylated and further metabolized.40 Mammalian cells do not have transporters for this sugar.41 The first approach to radiolabel a sorbitol analog was done by Li et al. who used a 30-min reaction to reduce 18F-FDG into 18F-fluorodeoxysorbitol (18F-FDS).42 Subsequently, Weinstein and Ordonez et al. demonstrated that 18F-FDS rapidly and selectively accumulated in Enterobacteriaceae (~1000 fold higher than mammalian cells), including MDR clinical strains, but not in Gram-positive bacteria or mammalian/cancer cells (Fig. 2).43 In a murine myositis model, 18F-FDS PET rapidly differentiated infection from sterile inflammation with a limit of detection of 6.2 ± 0.2 log10 colony forming units (CFU) for E. coli. This is a promising threshold since clinically relevant infections have much higher burdens of approximately 8 log10 CFU bacteria per mL,44 with volumes of tens to hundreds of milliliters.45,46 These findings have been extended to models of mixed Gram-positive and Gram-negative thigh co-infections, brain infection, mice undergoing immunosuppressive chemotherapy, and K. pneumoniae pneumonia.43,47,48 Importantly, 18F-FDS PET also monitored the efficacy of antimicrobial treatment with the PET signal correlating with the bacterial burden. Recently, 18F-FDS was determined to be safe and well tolerated after a single intravenous dose injected into healthy human volunteers to assess biodistribution and radiation dosimetry.49

Figure 2. 18F-FDS PET/CT imaging of E. coli myositis in immunocompetent mice.

Figure 2

18F-FDS signal is noted in the infected (yellow arrow) but not in the inflamed (control) sterile thigh (red arrow). 18F-FDG signal was noted in both infected and inflamed thighs. Reprinted with permission by the American Association for the Advancement of Science from Weinstein and Ordonez et al. 2014.43

Maltose and maltodextrin are polysaccharides consisting of two or multiple glucose units respectively. These sugars are incorporated with high specificity using the maltose-maltodextrin transport system, present in multiple Gram-negative and Gram-positive bacterial species, but not in mammalian cells.50 In 2011, Ning et al. reported maltodextrin-based optical imaging probes that could specifically-detect E. coli myositis in mice with high specificity.51 Subsequent work led to the development of 18F-labeled maltohexaose (MH18F).52 Using 19F NMR spectroscopy, MH19F was found to have high specificity for bacteria over mammalian cells, accumulating two times more in E. coli compared to hepatocytes. Using a rat E. coli myositis model, MH18F PET was able to localize the infected tissue as early as 10 min post-injection, and 60 min later the signal in infected muscle was 8.5-fold higher compared to uninfected control muscle. Unlike 18F-FDG, MH18F was able to differentiate between live (infection) and dead (inflammation) bacteria in muscle. MH18F was also used to effectively measure drug resistance and monitor the therapeutic effect of antibiotics in vivo. Therefore, MH18F could be a useful PET imaging agent for a broad spectrum of Gram-positive and Gram-negative infections. A similar approach was adopted by Gowrishankar et al. in the development of 6-18F-fluoromaltose, which accumulated in in vitro cultures of E. coli, P. aeruginosa and L. monocytogenes but not in mammalian cells.53 PET imaging with this compound was also able to differentiate infected from inflamed muscle. However, 6-18F-fluoromaltose had suboptimal pharmacokinetics and poor signal-to-background ratios, leading to the development of the second-generation tracer 6″-18F-fluoromaltotriose, with promising results detecting E. coli myositis and P. aeruginosa wound infections in vivo (Fig. 3).54

Figure 3. 6″-18F-fluoromaltotriose PET/CT imaging of E. coli myositis.

Figure 3

Three-dimensional color map from small-animal PET/CT scan of representative nude mouse with E. coli infection in its right thigh 24 h after infection and 1 h after intravenous injection of 7.4 MBq of 6″-18F-fluoromaltotriose. Also seen in image is bladder (BL) and kidneys (K). Yellow arrow highlights site of infection. This research was originally published in JNM. Gowrishankar, G. et al. J Nucl Med. 2017;58:1679–1684. © by the Society of Nuclear Medicine and Molecular Imaging, Inc.54

Trehalose is a disaccharide essential for mycobacterial growth and virulence55, and there is evidence of the uptake of trehalose analogs by Mycobacterium tuberculosis.56 Recently, Rundell et al. reported that a fluorinated trehalose analog (19F-FDTre) accumulates in Mycobacterium smegmatis and provided a synthetic scheme for a 18F-radiolabeled version.57 Although these results are still preliminary, they suggest the potential of radiolabeled trehalose analogs for Mycobacteria-specific imaging.

Cell wall components

Amino sugars are essential components of peptidoglycan, the strong polymer of the bacterial cell wall that maintains the cell shape and anchors components of the cell envelope. The peptidoglycan is built from alternating units of N-acetylglucosamine and N-acetylmuramic acid, cross-linked with oligopeptides at the lactic acid residue of N-acetylmuramic acid.58 Using a N-acetylglucosamine derivative, Martinez et al. reported the synthesis, preliminary characterization, and PET imaging of 2-deoxy-2-18F-fluoroacetamido-D-glucopyranose (18F-FAG) in E. coli infected rats.59 The signal of 18F-FAG PET in infected muscle was 1.68 times higher compared to turpentine oil-induced inflammation. Another component of the peptidoglycan layer of the bacterial cell wall in both Gram-positive and Gram-negative bacteria are D-amino acids.60,61 Unlike mammalian cells, most bacteria incorporate and produce significant amounts of D-amino acids into their cell walls,62 being the main target of a large number of antibiotics that antagonize the synthesis, dimerization and/or incorporation of D-amino acids. Using this approach, Neumann et al. recently reported the radio-synthesis of D-methyl-11C-Methionine (11C-D-Met), demonstrating rapid and selective differentiation of both E. coli and S. aureus from sterile inflammation in a myositis mouse model (Fig. 4).63 Conversely, L-methyl-11C-Methionine is not specific for infection imaging.63,64 Preliminary studies have considered the potential of bacterial wall-labeling agents such as D-Alanine and D-Glutamate. However, significant in vivo defluorination for 18F-labeled versions of alanine,65,66 modest enantiomeric excess for the reported 11C-L-Alanine asymmetric synthesis,67 and the lack of homology between reported 18F versions of glutamate and the native amino acid are significant drawbacks.68,69 Previous studies have also indicated that many unnatural D-amino acids are incorporated into bacterial peptidoglycan with a high tolerance for structural differences, suggesting the existence of potential bacteria-specific imaging compounds that are yet to be explored.70,71

Figure 4. In vivo studies using 11C-D-Met in a murine myositis model.

Figure 4

Representative images show marked uptake in areas corresponding to live bacterial injection (left deltoid), in contrast to sterile inflammation (right deltoid) and normal muscle. The site of live bacterial inoculation is denoted by a red arrow, while the site of 10X heat-killed bacterial inoculation is denoted by a white arrow. Reprinted with permission by Springer Nature from Neumann et al. 2017.63

Iron metabolism

For more than 40 years, 67Ga-citrate SPECT has been extensively used to detect infection/inflammation.29,72 Multiple pathways have been proposed for 67Ga-citrate uptake in inflamed/infected tissues such as increased vascular permeability in the infection site,73 direct uptake by bacteria,74 uptake by leukocytes,75 and binding to plasma transferrin, lactoferrin and/or bacterial siderophores.76,77 Despite not being specific for bacterial infection, 67Ga-citrate is still widely used around the world to identify sites of infection in patients with FUO.78 Due to its shorter half-life, 68Ga-citrate was developed to improve the long waiting time required for imaging after a 67Ga-citrate injection.76 68Ga-citrate has been evaluated in animal models of osteomyelitis and myositis 79,80, as well as in patients with osteomyelitis.81 In patients with tuberculosis, 68Ga-citrate accumulated in both pulmonary and extra-pulmonary tuberculous lesions.82 Based on the same principle of iron accumulation in infected tissues, Kumar et al. developed a 68Ga-radiolabeled apo-transferrin complex (68Ga-TF), which provided 7.5 times higher signal in the PET images of S. aureus muscle infection in rats, compared to uninfected uninflamed muscle.83 No signal from the infected sites was evident within the first 2-hour post-injection in additional unconjugated 68Ga-Cl3 PET scans, suggesting some specificity of the transferrin-bound compound. Although these mechanisms are not bacteria-specific, there has also been interest in the use of pathogen-specific radiolabeled siderophores as potential bacteria-specific imaging agents.84

ANTIMICROBIAL PEPTIDES

Peptides are short chains of amino acids linked by amide bonds. Antimicrobial peptides are synthesized by cells involved in the host immune response, and due to their cationic properties, they tend to bind more to the negatively charged bacterial cell wall compared to eukaryotic cells.85,86 Based on the initial experiments with ubiquicidin (UBI), a human antimicrobial peptide present in the respiratory epithelium, Welling et al. synthesized and evaluated several 99mTc-radiolabeled fragments of UBI with increased sensitivity and selectivity for bacteria.87 Subsequent studies with 99mTc-UBI (fragments 29–41) showed promising results imaging Gram-positive and Gram-negative bacteria, including MDR bacterial strains, and monitoring antimicrobial efficacy in both preclinical and clinical settings.8893 Based on the same peptide, Welling et al. developed 111In-DTPA-Cy5-UBI(29–41), a hybrid fluorophore and radionuclide labeled antimicrobial peptide, which had a 2.82 and 2.37 target to background ratio in a S. aureus and K. pneumoniae myositis mouse model, respectively.94 18F-UBI(29–41) has also been evaluated in a rat S. aureus myositis model with poor results due to extensive defluorination.95 In 2014, Ebenhan et al. synthesized 68Ga-NOTA-UBI(29–41) and showed specificity towards S. aureus in a rabbit model, being able to distinguish infection from sterile inflammation.96 These findings were subsequently validated in a S. aureus mouse myositis model.97 The first-in-human study evaluating the diagnostic performance of 68Ga-NOTA-UBI(29–41) was performed in three patients with known/suspected bone-or soft tissue infection.98 With a target to background ratio of 3.4 ± 0.2 at 60 minutes, 68Ga-NOTA-UBI(29–41) was able to accurately identify the infection sites in these patients (Fig. 5). No adverse effects were reported with the administration of 68Ga-NOTA-UBI(29–41).

Figure 5. 68Ga-NOTA-UBI(29–41) image in a patient with peripheral bone and soft-tissue infection.

Figure 5

(A) Whole-body MIP-PET image shows a diffusely increased tracer uptake in the right lower leg. (B) Detailed PET-CT images demonstrate focal increased tracer uptake in the ankle-joint extending in the adjacent bone (black and white arrows) as well as diffuse tracer uptake in the calf muscles (red arrow) whereas there is no significant uptake in the contralateral leg. Images are obtained 60 min after tracer administration (240 MBq). This research was originally published in JNM. Ebenhan, T. et al. J Nucl Med. 2017. © by the Society of Nuclear Medicine and Molecular Imaging, Inc.98

Using a derivative of depsidomycin, a peptide with significant antimicrobial activity, Mokaleng et al. developed and evaluated 68Ga-DOTA-TBIA101 in a mouse model of E. coli myositis, where the tracer was able to differentiate infection from healthy tissue.99 However, this same peptide accumulated in rabbit muscles infected with M. tuberculosis and sterile inflammation controls at the same rate, suggesting a non-specific uptake by the inflammation associated with the infection rather than by the bacteria.100 In 2017, Dutta et al. radiolabeled and reported LL37, a human cathelicidin antimicrobial peptide (68Ga-CDP1) that had higher uptake in in vitro bacterial cultures of S. aureus, E. coli and M. smegmatis, compared to eukaryotic cells.101 Additional not bacteria-specific radiolabeled peptides such as the human neutrophil peptide (99mTc-HNP-1), neutrophil elastase inhibitor peptide (99mTc-HNE-2), human β-defensin (99mTc-HBD-3), and vascular adhesion protein-1 selective peptide (68Ga-DOTAVAP-P1), have been evaluated for bacterial imaging with variable results.102105

ANTIBIOTICS

Antibiotics are compounds designed to kill (bactericidal) or impair the growth (bacteriostatic) of bacterial pathogens. Ideally, a radiolabeled antibiotic will bind to their bacterial targets with high specificity. However, an excess of unlabeled antibiotic may compete with the radiolabeled antibiotic or kill the pathogen, both of which will impair the imaging efficacy of the tracer. Additionally, desirable pharmacologic characteristics of antibiotics may not match those for radiolabeled bacterial imaging compounds (e.g. long half-life for convenient daily dosing). Moreover, bacterial resistance mechanisms to a specific antibiotic, such as efflux pumps, cell permeability changes, decreased target affinity or degradative metabolism can affect the radiolabeled analog and could limit its use as a diagnostic tracer. Radiolabeled antibiotics and PET imaging also provide a noninvasive technique for studying pharmacokinetics and pharmacodynamics in vivo.106110 A large number of antibiotics have been radiolabeled, with 99mTc being the predominant radionuclide since the tracer preparation is easily accomplished using a cold kit format within 30 minutes.111113 However, a large number of these radiolabeled antibiotics have not been able to differentiate infection from inflammation and have not been evaluated in a clinical setting. While we provide a brief discussion of the most significant radiolabeled antibiotics, multiple extensive reviews are available on this topic.106,114117

Fluoroquinolones

Ciprofloxacin, a broad-spectrum bactericidal fluoroquinolone, was initially labeled with 99mTc and evaluated in preclinical infection models with promising results.118 99mTc-Ciprofloxacin imaging was used in large clinical trials that reported high sensitivity and specificity in the diagnosis of bacterial infections.119,120 However, subsequent preclinical experiments and clinical trials with 99mTc-ciprofloxacin or 18F-ciprofloxacin demonstrated variable specificity and an inability to reliably differentiate infection from sterile inflammatory processes.121124 Development of this tracer proved controversial due to low specificity attributed to nonspecific binding to dead bacteria,125 as well as nonspecific binding interactions with host neutrophil DNA.126 Commercial development of 99mTc-ciprofloxacin was suspended after an unsuccessful phase II trial in which it failed to differentiate sterile inflammation from infection.127 Nevertheless, 99mTc-ciprofloxacin has continued to be used by some investigators for the diagnosis of infections.128,129 Multiple additional fluoroquinolones, including lomefloxacin,130 fleroxacin131 and trovafloxacin,132 have been radiolabeled with 18F and/or 99mTc, but preliminary studies have demonstrated inconsistent findings.106,114 Although some have promising results for bacteria-specific imaging, extensive preclinical evaluation for the uptake mechanisms should be performed before further development in order to avoid past mistakes.

Cephalosporins

As a class of β-lactam antibiotics that directly targets the cell wall, multiple cephalosporins have been radiolabeled and evaluated for bacterial imaging. Initial experiments with 99mTc-ceftriaxone in animal models (mice, rats, and rabbits) suggested higher accumulation in infected versus inflamed tissues.133135 Preliminary studies in humans with 99mTc-ceftriaxone showed promising results with 80% sensitivity to detect orthopedic infections.136 In 2012, Kaul et al. reported SPECT imaging with 99mTc-ceftriaxone in 36 patients with clinical and radiological suspicion of an orthopedic infection. They reported a sensitivity and specificity of 85.2% and 77.8%, respectively, for the diagnosis of infection.137 The high protein binding of 99mTc-ceftriaxone (>90%) led to delayed elimination, and a high background. Additional preclinical experiments have been done with other cephalosporins such as 99mTc-cefepime, which demonstrated selective accumulation in an E. coli-infected thigh muscle compared to a heat-killed E. coli and a turpentine oil induced inflammation in a rat model.138 Similar experiments have been reported for 99mTc-labeled cefazolin,139 cefoperazone,140 cefotaxime,141 ceftazidime,142 ceftizoxime143 and cefuroxime.144

Antifolates

Based on the structure of trimethoprim, a synthetic antibiotic that inhibits the bacterial dihydrofolate reductase, an enzyme conserved in most bacteria that is involved in DNA synthesis and the folate pathway, Sellmyer et al. developed 18F-fluoropropyl-trimethoprim (18F-FPTMP), showing in vitro accumulation in S. aureus, E. coli and P. aeruginosa.145 In a mouse myositis model, 18F-FPTMP identified sites of bacterial infection with E. coli and S. aureus and differentiated infection from sterile inflammation and tumor (Fig. 6). The authors also evaluated the biodistribution of 18F-FPTMP in non-infected non-human primates with promising results for its translation to humans.

Figure 6. 18F-FPTMP PET/CT imaging of E. coli myositis.

Figure 6

A mouse infected live E. coli (8 log10 CFU injected into the right lower leg) was imaged after injection with 18F-FPTMP, ~7.4 MBq IV. There is uptake in the infected hind limb muscle (arrow) 4 h after infection, but not in the area of turpentine injection (arrowhead). Next-day imaging with 18F-FDG, ~11.1 MBq IV, shows uptake in both infection and chemical inflammation 1 h after injection. Reprinted with permission by the National Academy of Sciences of the USA from Sellmyer et al. 2017.145

Several other radiolabeled antibiotics with different mechanisms of action have been evaluated.106,114 However, the differences between the experimental designs, makes the comparison amongst them difficult. The variability between animal models (mice, rats or rabbits), injected activities and image acquisition times are significant, and, most importantly, the number of bacteria used for inducing the infection ranges from 5 to 10 log10 colony forming units (CFU).114 Moreover, given the rapid growth of bacteria in tissues (generation time ~20 min for most pyogenic bacteria), information on the bacterial burden at the time of imaging/radioactivity quantification rather than at the time of injection would be the critical determinant of sensitivity.

LEUKOCYTES/ANTIBODIES

In vitro leukocytes, labeled with 111In (111In-oxine-leukocyte) or 99mTc (99mTc-HMPAO-leukocyte), remains one of the most commonly clinically performed molecular imaging test for most infections in the immunocompetent population.146 However, in vitro labeling of leukocytes has significant disadvantages since the labeling procedure is labor intensive, involves contact with blood products and may be difficult to perform in leukopenic patients, since it depends on intact chemotaxis, the number and types of cells labeled, and the cellular response to the specific infection.29,147 Furthermore, since most of the radiolabeled cells are neutrophils, leukocyte imaging is much less sensitive in infections that do not elicit a predominantly neutrophilic cellular response, such as tuberculosis.148

Multiple radiolabeled antibodies (e.g. 99mTc-besilesomab, 111In-IgG) and antibody fragments (e.g. 99mTc-sulesomab) have been developed and evaluated with promising results.111,149 Besilesomab, a murine-derived monoclonal antibody of the IgG1 κ isotype, radiolabeled with 99mTc has been used with variable results in the evaluation of FUO, joint infection and osteomyelitis.150152 99mTc-sulesomab is a 50 kDa fragment antigen-binding (Fab) portion of an IgG1 class murine monoclonal antibody that binds to the normal cross-reactive antigen-90 (NCA-90) present on leukocytes. Although sensitive for diagnosis of soft tissue infections, 99mTc-sulesomab is not specific for bacteria.153,154

While most of these compounds target the inflammation associated with infection, antibodies specifically targeting bacteria have also been reported, although more work is needed to improve the several pharmacokinetic challenges including slow clearance from non-target sites. Early research with a 99mTc-labeled anti-S. aureus antibody showed encouraging results in the rabbit endocarditis model.155 Using a similar endocarditis model in rats, Pinkston et al. reported a monoclonal antibody (MAb 69) against the major component of Enterococcus faecalis pili, EbpC.156 After radiolabeling, 64Cu-DOTA-MAb 69 PET imaging successfully identified E. faecalis endocarditis. Targeting a different pathogen, in 1988 Rubin et al. reported a 125I-labeled anti-P. aeruginosa specific monoclonal antibody that was able to discriminate between P. aeruginosa and S. aureus infection sites in a rat model.157 Recently, Wiehr et al. developed a 64Cu-labeled polyclonal antibody targeting YadA, an outer membrane protein, essential for in vivo virulence of Yersinia enterocolitica.158 64Cu-NODAGA-YadA PET allowed the detection of bacteria in the spleen, although a clear identification of the pathogen in the liver was not possible due to the nonspecific uptake of the radiolabeled antibody and free-copper accumulation. Using a similar approach and the similarities between M. tuberculosis and Mycobacterium bovis (Bacillus Calmette–Guérin, BCG), Malpani et al. developed a 131I-labeled anti-BCG antibody.159 Localization of 131I-anti BCG was observed at the site of TB lesions in a rabbit model, but significant concentration of 131I-anti BCG was also noted in the background and major organs like heart, liver, spleen, and kidneys. Subsequently, Lee et al. reported the use of a 131I-labeled antibody against BCG,160 demonstrating that the BCG-specific antibody F(ab′)2 accumulated in TB lesions of rabbits (induced with by the inoculation of heat-killed, sonicated M. tuberculosis in muscle) while washing away from syphilitic lesions. However, it is possible that the results could be attributed to non-specific iodine uptake in the inflammation site.161

APTAMERS/OLIGOMERS

Aptamers are single-stranded oligonucleotides with high affinity and specificity to the target molecules, ideal characteristics for a bacteria-specific imaging tracer.162 Most of the work with aptamers for bacteria-imaging has been based on optical imaging probes.163 In 2015, S. aureus-specific aptamers SA20, SA23 and SA34 were radiolabeled with 99mTc and showed high uptake in the infection site compared to background.164 Recently, using aptamers designed to bind to the peptidoglycan of Gram-negative and Gram-positive bacteria, Ferreira et al. recently described 99mTc-Antibac1 and showed it could localize S. aureus infection in a mouse model although it has high binding to plasma proteins, which could limit its use.165 Using a similar approach, Chen et al. synthesized a radiolabeled oligomer complementary to the bacterial 16S rRNA (99mTc-MORF), and evaluated its accumulation in a K. pneumoniae infection in a myositis mouse model.166 A more detailed review of the use of aptamers and oligomers for bacterial imaging is also available.167

BACTERIOPHAGES

Bacteriophages are viruses that show no specificity for mammalian cells and infect bacteria exclusively by attaching to specific surface receptors and insert their genetic material into the cell to use it as a host for reproduction.168 This approach was initially used by Rusckowski et al. who reported that the 99mTc-labeled bacteriophage M13 had a 1.6 times higher activity in the E. coli or S. aureus-infected mouse thigh compared to inflamed muscle.169 A subsequent publication from the same group described the bacteriophages P22, E79, VD-13 and 60, labeled with 99mTc using MAG(3) as the chelator. The P. aeruginosa-specific bacteriophage 99mTc-E79 had promising results being able to differentiate infection from inflammation in a myositis mouse model.170 More recently, Cardoso et al. evaluated another 99mTc-labeled P. aeruginosa-specific bacteriophage (PP7), describing a 1.68 times higher signal in the infected side compared with sterile inflammation.171 In these studies, the authors describe positive signal from the gastrointestinal tract, suggesting that the bacteriophages are binding to the host microbiome.

VITAMINS

Biotin (vitamin B7) is a precursor of acetyl CoA carboxylase in bacteria and eukaryotic cells.172 111In-labeled biotin, alone or combined with streptavidin, has been reported for bacterial SPECT imaging in patients and animal models,173175 with promising results in the diagnosis of vertebral osteomyelitis. However, this approach is unlikely to be bacteria-specific since streptavidin accumulates at the infection site as well as the surrounding inflammation.176 Additional biotin analogs have also been radiolabeled with 18F and evaluated in E. coli-infected rats.177 Recently, Baldoni et al. reported an analog of cyanocobalamin (vitamin B12) labeled with 99mTc, 99mTc-PAMA(4)-Cbl.178 This tracer had a time-dependent accumulation in vitro in S. aureus and E. coli, and ex vivo tissue biodistribution showed a higher concentration of tracer in the infected implants.

OTHER AGENTS

The negatively-charged phospholipids in bacterial membranes bind to bis(zinc(II)-dipicolylamine) (Zn-DPA). This approach was used with fluorescent Zn-DPA that accumulated in vitro in Gram-positive and Gram-negative bacteria, using optical imaging to monitor in vivo imaging of infection mouse models.179,180 Subsequently, Liu et al. developed 111In-DOTA-biotin and Zn-DPA-biotin, non-covalently linked by streptavidin to form the complex 111In-DOTA-biotin-SA-Zn-DPA-biotin.181 Using a myositis mouse model, the signal of the 111In-labeled Zn-DPA complex was 2.8-fold higher in the infected animals compared to inflamed controls. Using a different chelator, Rice et al. used 111In-DTPA-Zn-DPA SPECT to successfully identify the Streptococcus pyogenes infection in the mouse model.182 However, since dead mammalian cells have an increased negative charge on the cell wall, Zn-DPA was found to also bind to necrotic and apoptotic tissues reducing its specificity for bacteria.183 99mTc and 18F-labeled analogs of Zn-DPA have been reported to successfully evaluate death and cardiomyocyte apoptosis following acute myocardial infarction.184186

A different approach targeting a bacterial enzyme was reported by Panizzi et al. During infection with coagulase-positive Staphylococci (e.g. S. aureus), prothrombin binds to staphylocoagulase, a fibrinogen-binding protein, to form an active complex that has fibrinogen-clotting capabilities. Using a small inhibitory peptide that binds to prothrombin for radiolabeling, PET/CT imaging with 64Cu-DTPA-prothrombin (64Cu-DTPA-ProT) was able to detect S. aureus valve vegetations in a mouse model of endocarditis.187

CONCLUSION

For years, non-specific tracers that target infection-associated inflammation have been widely used in clinical practice. Their application has led to early detection of infectious diseases, deeper understanding of bacterial pathogenesis, antibiotic therapy decision making and treatment follow up. However, in the last decade, increasing interest and research efforts have been invested in search of specific probes for imaging bacterial infections. The ability to rapidly and non-invasively discriminate between infectious and sterile inflammation and the potential of differentiating between bacterial classes (and species) will prove to be very valuable for the practicing clinician. Although most of the reported bacteria-specific tracers are in preclinical stages, some are rapidly moving to the clinical setting with promising results. The next couple of years promise to be an exciting period for bacterial-specific imaging, as the search for the ideal tracers continues to be a challenge, but the goal does not seem impossible to achieve anymore.

Acknowledgments

This study was funded by the National Institutes of Health (NIH) Director’s Transformative Research Award R01-EB020539 (S.K.J.) and R01-HL131829 (S.K.J.). The funders had no role in study design, data collection, and analysis, decision to publish, or preparation of the manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Antibiotic resistance threats in the United States, 2013. Centres for Disease Control and Prevention, US Department of Health and Human Services; 2013. [Google Scholar]
  • 2.McCaughey B. [accessed November 1, 2017];Unnecessary Deaths: The Human and Financial Costs of Hospital Infections. (2). http://emerald.tufts.edu/med/apua/consumers/faqs_2_4154863510.pdf.
  • 3.Scott RD. [accessed November 1, 2017];The Direct Medical Costs of Healthcare-Associated Infections in U.S. Hospitals and the Benefits of Prevention. 2009 http://www.cdc.gov/hai/pdfs/hai/scott_costpaper.pdf.
  • 4.Velmahos GC, Jindal A, Chan L, et al. Prophylactic antibiotics after severe trauma: more is not better. Int Surg. 2001;86(3):176–83. [PubMed] [Google Scholar]
  • 5.Velmahos GC, Toutouzas KG, Sarkisyan G, et al. Severe trauma is not an excuse for prolonged antibiotic prophylaxis. Arch Surg. 2002;137(5):537–41. doi: 10.1001/archsurg.137.5.537. discussion 41–2. [DOI] [PubMed] [Google Scholar]
  • 6.McKenna M. Antibiotic resistance: the last resort. Nature. 2013;499(7459):394–6. doi: 10.1038/499394a. [DOI] [PubMed] [Google Scholar]
  • 7.Nordmann P, Dortet L, Poirel L. Carbapenem resistance in Enterobacteriaceae: here is the storm! Trends in molecular medicine. 2012;18(5):263–72. doi: 10.1016/j.molmed.2012.03.003. [DOI] [PubMed] [Google Scholar]
  • 8.Salazar-Austin N, Ordonez AA, Hsu AJ, et al. Extensively drug-resistant tuberculosis in a young child after travel to India. Lancet Infect Dis. 2015;15(12):1485–91. doi: 10.1016/S1473-3099(15)00356-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lungren MP, Peper JPC, Ordoñez AA, Jain SK. Infectious Disease Imaging. In: Mollura DJ, Lungren MP, editors. Radiology in Global Health: Strategies, Implementation, and Applications. New York, NY: Springer New York; 2014. pp. 159–79. [Google Scholar]
  • 10.Heuker M, Gomes A, van Dijl JM, et al. Preclinical studies and prospective clinical applications for bacteria-targeted imaging: the future is bright. Clinical and Translational Imaging. 2016;4(4):253–64. doi: 10.1007/s40336-016-0190-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Archer NK, Francis KP, Miller LS. Optical Imaging. In: Jain SK, editor. Imaging Infections: From Bench to Bedside. Cham: Springer International Publishing; 2017. pp. 43–76. [Google Scholar]
  • 12.Zackrisson S, van de Ven SM, Gambhir SS. Light in and sound out: emerging translational strategies for photoacoustic imaging. Cancer Res. 2014;74(4):979–1004. doi: 10.1158/0008-5472.CAN-13-2387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mills B, Bradley M, Dhaliwal K. Optical imaging of bacterial infections. Clinical and translational imaging. 2016;4(3):163–74. doi: 10.1007/s40336-016-0180-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bardhan NM, Ghosh D, Belcher AM. Carbon nanotubes as in vivo bacterial probes. Nature communications. 2014;5:4918. doi: 10.1038/ncomms5918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jain SK. The Promise of Molecular Imaging in the Study and Treatment of Infectious Diseases. Molecular Imaging and Biology. 2017;19(3):341–7. doi: 10.1007/s11307-017-1055-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Palestro CJ. Radionuclide imaging of infection: in search of the grail. Journal of Nuclear Medicine. 2009;50(5):671–3. doi: 10.2967/jnumed.108.058297. [DOI] [PubMed] [Google Scholar]
  • 17.Smith G, Carroll L, Aboagye EO. New frontiers in the design and synthesis of imaging probes for PET oncology: current challenges and future directions. Molecular imaging and biology. 2012;14(6):653–66. doi: 10.1007/s11307-012-0590-y. [DOI] [PubMed] [Google Scholar]
  • 18.Johnson DH, Via LE, Kim P, et al. Nuclear imaging: a powerful novel approach for tuberculosis. Nuclear medicine and biology. 2014;41(10):777–84. doi: 10.1016/j.nucmedbio.2014.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gemmel F, Dumarey N, Welling M. Seminars in nuclear medicine. Vol. 2009. Elsevier; 2009. Future diagnostic agents; pp. 11–26. [DOI] [PubMed] [Google Scholar]
  • 20.Pysz MA, Gambhir SS, Willmann JK. Molecular imaging: current status and emerging strategies. Clinical radiology. 2010;65(7):500–16. doi: 10.1016/j.crad.2010.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mac Faddin JF. Biochemical tests for identification of medical bacteria. Baltimore, MD: The Williams & Wilkins Company; 1976. [Google Scholar]
  • 22.Ordonez AA, Weinstein EA, Bambarger LE, et al. A systematic approach for developing bacteria-specific imaging tracers. Journal of Nuclear Medicine. 2017;58(1):144–50. doi: 10.2967/jnumed.116.181792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Peterson KL, Reid WC, Freeman AF, et al. The use of 14 C-FIAU to predict bacterial thymidine kinase presence: implications for radiolabeled FIAU bacterial imaging. Nuclear medicine and biology. 2013;40(5):638–42. doi: 10.1016/j.nucmedbio.2013.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bettegowda C, Foss CA, Cheong I, et al. Imaging bacterial infections with radiolabeled 1-(2′-deoxy-2′-fluoro-beta-D-arabinofuranosyl)-5-iodouracil. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(4):1145–50. doi: 10.1073/pnas.0408861102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pullambhatla M, Tessier J, Beck G, Jedynak B, Wurthner JU, Pomper MG. [(125)I]FIAU imaging in a preclinical model of lung infection: quantification of bacterial load. Am J Nucl Med Mol Imaging. 2012;2(3):260–70. [PMC free article] [PubMed] [Google Scholar]
  • 26.Diaz LA, Jr, Foss CA, Thornton K, et al. Imaging of musculoskeletal bacterial infections by [124I]FIAU-PET/CT. PLoS One. 2007;2(10):e1007. doi: 10.1371/journal.pone.0001007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhang XM, Zhang HH, McLeroth P, et al. [124I]FIAU: Human dosimetry and infection imaging in patients with suspected prosthetic joint infection. Nuclear Medicine and Biology. 2016;43(5):273–9. doi: 10.1016/j.nucmedbio.2016.01.004. [DOI] [PubMed] [Google Scholar]
  • 28.Nimmagadda S, Mangner TJ, Douglas KA, Muzik O, Shields AF. Biodistribution, PET, and Radiation Dosimetry Estimates of HSV-tk Gene Expression Imaging Agent 1-(2′-Deoxy-2′-18F-Fluoro-β-d-Arabinofuranosyl)-5-Iodouracil in Normal Dogs. Journal of Nuclear Medicine. 2007;48(4):655–60. doi: 10.2967/jnumed.106.036830. [DOI] [PubMed] [Google Scholar]
  • 29.Chen DL, Scherer PM, Palestro CJ. Clinical Imaging. In: Jain SK, editor. Imaging Infections: From Bench to Bedside. Cham: Springer International Publishing; 2017. pp. 17–41. [Google Scholar]
  • 30.Lawal I, Zeevaart JR, Ebenhan T, et al. Metabolic Imaging of Infection. Journal of Nuclear Medicine. 2017 doi: 10.2967/jnumed.117.191635. jnumed. 117.191635. [DOI] [PubMed] [Google Scholar]
  • 31.Vaidyanathan S, Patel CN, Scarsbrook AF, Chowdhury FU. FDG PET/CT in infection and inflammation--current and emerging clinical applications. Clin Radiol. 2015;70(7):787–800. doi: 10.1016/j.crad.2015.03.010. [DOI] [PubMed] [Google Scholar]
  • 32.Marjanovic S, Skog S, Heiden T, Tribukait B, Nelson BD. Expression of glycolytic isoenzymes in activated human peripheral lymphocytes: cell cycle analysis using flow cytometry. Exp Cell Res. 1991;193(2):425–31. doi: 10.1016/0014-4827(91)90116-c. [DOI] [PubMed] [Google Scholar]
  • 33.Jamar F, Buscombe J, Chiti A, et al. EANM/SNMMI guideline for 18F-FDG use in inflammation and infection. J Nucl Med. 2013;54(4):647–58. doi: 10.2967/jnumed.112.112524. [DOI] [PubMed] [Google Scholar]
  • 34.Chen RY, Dodd LE, Lee M, et al. PET/CT imaging correlates with treatment outcome in patients with multidrug-resistant tuberculosis. Science translational medicine. 2014;6(265):265ra166. doi: 10.1126/scitranslmed.3009501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Stelzmueller I, Huber H, Wunn R, et al. 18F-FDG PET/CT in the Initial Assessment and for Follow-up in Patients With Tuberculosis. Clinical nuclear medicine. 2016;41(4):e187–e94. doi: 10.1097/RLU.0000000000001102. [DOI] [PubMed] [Google Scholar]
  • 36.Granados U, Fuster D, Pericas JM, et al. Diagnostic accuracy of 18F-FDG PET/CT in infective endocarditis and implantable cardiac electronic device infection: a cross-sectional study. Journal of Nuclear Medicine. 2016;57(11):1726–32. doi: 10.2967/jnumed.116.173690. [DOI] [PubMed] [Google Scholar]
  • 37.Love C, Marwin SE, Palestro CJ. Nuclear medicine and the infected joint replacement. Semin Nucl Med. 2009;39(1):66–78. doi: 10.1053/j.semnuclmed.2008.08.007. [DOI] [PubMed] [Google Scholar]
  • 38.Treglia G, Sadeghi R, Annunziata S, et al. Diagnostic performance of Fluorine-18-Fluorodeoxyglucose positron emission tomography for the diagnosis of osteomyelitis related to diabetic foot: a systematic review and a meta-analysis. The Foot. 2013;23(4):140–8. doi: 10.1016/j.foot.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 39.Mills B, Awais RO, Luckett J, et al. [(18)F]FDG-6-P as a novel in vivo tool for imaging staphylococcal infections. EJNMMI Res. 2015;5:13. doi: 10.1186/s13550-015-0095-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lengeler J. Nature and properties of hexitol transport systems in Escherichia coli. J Bacteriol. 1975;124(1):39–47. doi: 10.1128/jb.124.1.39-47.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Scott ME, Viola RE. The use of fluoro-and deoxy-substrate analogs to examine binding specificity and catalysis in the enzymes of the sorbitol pathway. Carbohydrate research. 1998;313(3):247–53. doi: 10.1016/s0008-6215(98)00266-3. [DOI] [PubMed] [Google Scholar]
  • 42.Li Z-B, Wu Z, Cao Q, et al. The synthesis of 18F-FDS and its potential application in molecular imaging. Molecular Imaging and Biology. 2008;10(2):92–8. doi: 10.1007/s11307-007-0125-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Weinstein EA, Ordonez AA, DeMarco VP, et al. Imaging Enterobacteriaceae infection in vivo with 18F-fluorodeoxysorbitol positron emission tomography. Science translational medicine. 2014;6(259):259ra146–259ra146. doi: 10.1126/scitranslmed.3009815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Konig C, Simmen HP, Blaser J. Bacterial concentrations in pus and infected peritoneal fluid--implications for bactericidal activity of antibiotics. J Antimicrob Chemother. 1998;42(2):227–32. doi: 10.1093/jac/42.2.227. [DOI] [PubMed] [Google Scholar]
  • 45.Jang K, Lee DH, Lee SH, Chung BH. Treatment of prostatic abscess: case collection and comparison of treatment methods. Korean J Urol. 2012;53(12):860–4. doi: 10.4111/kju.2012.53.12.860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yamamoto M, Fukushima T, Hirakawa K, Kimura H, Tomonaga M. Treatment of bacterial brain abscess by repeated aspiration--follow up by serial computed tomography. Neurol Med Chir (Tokyo) 2000;40(2):98–104. doi: 10.2176/nmc.40.98. discussion -5. [DOI] [PubMed] [Google Scholar]
  • 47.Li J, Zheng H, Fodah RA, Warawa J, Ng CK. Validation of 2–18F-fluorodeoxysorbitol (18F-FDS) as a potential radiopharmaceutical for imaging bacterial infection in the lung. Journal of Nuclear Medicine. 2017 doi: 10.2967/jnumed.117.195420. [DOI] [PubMed] [Google Scholar]
  • 48.Yao S, Xing H, Zhu W, et al. Infection imaging with 18 F-FDS and first-in-human evaluation. Nuclear medicine and biology. 2016;43(3):206–14. doi: 10.1016/j.nucmedbio.2015.11.008. [DOI] [PubMed] [Google Scholar]
  • 49.Zhu W, Yao S, Xing H, et al. Biodistribution and Radiation Dosimetry of the Enterobacteriaceae-Specific Imaging Probe [18F]Fluorodeoxysorbitol Determined by PET/CT in Healthy Human Volunteers. Molecular Imaging and Biology. 2016;18(5):782–7. doi: 10.1007/s11307-016-0946-9. [DOI] [PubMed] [Google Scholar]
  • 50.Shuman HA, Treptow NA. The Maltose-Maltodextrin-Transport System of Escherichia coli K-12. In: Martonosi A, editor. The Enzymes of Biological Membranes. Springer Science & Business Media; 2012. pp. 561–72. [Google Scholar]
  • 51.Ning X, Lee S, Wang Z, et al. Maltodextrin-based imaging probes detect bacteria in vivo with high sensitivity and specificity. Nat Mater. 2011;10(8):602–7. doi: 10.1038/nmat3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ning X, Seo W, Lee S, et al. PET Imaging of Bacterial Infections with Fluorine-18-Labeled Maltohexaose. Angewandte Chemie International Edition. 2014;53(51):14096–101. doi: 10.1002/anie.201408533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gowrishankar G, Namavari M, Jouannot EB, et al. Investigation of 6-[18F]-Fluoromaltose as a Novel PET Tracer for Imaging Bacterial Infection. PLOS ONE. 2014;9(9):e107951. doi: 10.1371/journal.pone.0107951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gowrishankar G, Hardy J, Wardak M, et al. Specific Imaging of Bacterial Infection Using 6″-18F-Fluoromaltotriose: A Second-Generation PET Tracer Targeting the Maltodextrin Transporter in Bacteria. J Nucl Med. 2017;58(10):1679–84. doi: 10.2967/jnumed.117.191452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tournu H, Fiori A, Van Dijck P. Relevance of Trehalose in Pathogenicity: Some General Rules, Yet Many Exceptions. PLOS Pathogens. 2013;9(8):e1003447. doi: 10.1371/journal.ppat.1003447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Backus KM, Boshoff HI, Barry CS, et al. Uptake of unnatural trehalose analogs as a reporter for Mycobacterium tuberculosis. Nat Chem Biol. 2011;7(4):228–35. doi: 10.1038/nchembio.539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Rundell SR, Wagar ZL, Meints LM, et al. Deoxyfluoro-d-trehalose (FDTre) analogues as potential PET probes for imaging mycobacterial infection. Org Biomol Chem. 2016;14(36):8598–609. doi: 10.1039/c6ob01734g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Plumbridge J, Vimr E. Convergent pathways for utilization of the amino sugars N-acetylglucosamine, N-acetylmannosamine, and N-acetylneuraminic acid by Escherichia coli. Journal of bacteriology. 1999;181(1):47–54. doi: 10.1128/jb.181.1.47-54.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Martínez ME, Kiyono Y, Noriki S, et al. New radiosynthesis of 2-deoxy-2-[18F]fluoroacetamido-d-glucopyranose and its evaluation as a bacterial infections imaging agent. Nuclear Medicine and Biology. 2011;38(6):807–17. doi: 10.1016/j.nucmedbio.2011.02.006. [DOI] [PubMed] [Google Scholar]
  • 60.Kuru E, Hughes HV, Brown PJ, et al. In Situ probing of newly synthesized peptidoglycan in live bacteria with fluorescent D-amino acids. Angew Chem Int Ed Engl. 2012;51(50):12519–23. doi: 10.1002/anie.201206749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Caparros M, Pisabarro A, De Pedro M. Effect of D-amino acids on structure and synthesis of peptidoglycan in Escherichia coli. Journal of bacteriology. 1992;174(17):5549–59. doi: 10.1128/jb.174.17.5549-5559.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lam H, Oh D-C, Cava F, et al. D-amino acids govern stationary phase cell wall remodeling in bacteria. Science. 2009;325(5947):1552–5. doi: 10.1126/science.1178123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Neumann KD, Villanueva-Meyer JE, Mutch CA, et al. Imaging Active Infection in vivo Using D-Amino Acid Derived PET Radiotracers. Scientific Reports. 2017;7(1):7903. doi: 10.1038/s41598-017-08415-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.D’Souza MM, Sharma R, Jaimini A, et al. Metabolic assessment of intracranial tuberculomas using 11C-methionine and 18F-FDG PET/CT. Nuclear medicine communications. 2012;33(4):408–14. doi: 10.1097/MNM.0b013e32834f9b14. [DOI] [PubMed] [Google Scholar]
  • 65.Yang D, Kuang L-R, Cherif A, et al. Synthesis of [18F] fluoroalanine and [18F] fluorotamoxifen for imaging breast tumors. Journal of drug targeting. 1993;1(3):259–67. doi: 10.3109/10611869308996084. [DOI] [PubMed] [Google Scholar]
  • 66.Wang L, Zha Z, Qu W, et al. Synthesis and evaluation of 18 F labeled alanine derivatives as potential tumor imaging agents. Nuclear medicine and biology. 2012;39(7):933–43. doi: 10.1016/j.nucmedbio.2012.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fasth K-J, Antoni G, Langstrom B. Asymmetric synthesis of L-[3–11C]alanine and L-[3–11C]phenylalanine by a phase-transfer alkylation reaction. Journal of the Chemical Society, Perkin Transactions. 1988;1(12):3081–4. [Google Scholar]
  • 68.Krasikova RN, Kuznetsova OF, Fedorova OS, et al. 4-[18F] fluoroglutamic acid (BAY 85–8050), a new amino acid radiotracer for PET imaging of tumors: synthesis and in vitro characterization. Journal of medicinal chemistry. 2010;54(1):406–10. doi: 10.1021/jm101068q. [DOI] [PubMed] [Google Scholar]
  • 69.Koglin N, Mueller A, Berndt M, et al. Specific PET imaging of xC– transporter activity using a 18F-labeled glutamate derivative reveals a dominant pathway in tumor metabolism. Clinical Cancer Research. 2011;17(18):6000–11. doi: 10.1158/1078-0432.CCR-11-0687. [DOI] [PubMed] [Google Scholar]
  • 70.Siegrist MS, Whiteside S, Jewett JC, Aditham A, Cava F, Bertozzi CR. D-amino acid chemical reporters reveal peptidoglycan dynamics of an intracellular pathogen. ACS chemical biology. 2013;8(3):500–5. doi: 10.1021/cb3004995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pidgeon SE, Fura JM, Leon W, Birabaharan M, Vezenov D, Pires MM. Metabolic Profiling of Bacteria by Unnatural C- terminated D- Amino Acids. Angewandte Chemie. 2015;127(21):6256–60. doi: 10.1002/anie.201409927. [DOI] [PubMed] [Google Scholar]
  • 72.Littenberg RL, Taketa RM, Alazraki NP, Halpern SE, Ashburn WL. Gallium-67 for localization of septic lesions. Ann Intern Med. 1973;79(3):403–6. doi: 10.7326/0003-4819-79-3-403. [DOI] [PubMed] [Google Scholar]
  • 73.Tzen KY, Oster ZH, Wagner HN, Jr, Tsan MF. Role of iron-binding proteins and enhanced capillary permeability on the accumulation of gallium-67. J Nucl Med. 1980;21(1):31–5. [PubMed] [Google Scholar]
  • 74.Menon S, Wagner HN, Jr, Tsan MF. Studies on gallium accumulation in inflammatory lesions: II. Uptake by Staphylococcus aureus: concise communication. J Nucl Med. 1978;19(1):44–7. [PubMed] [Google Scholar]
  • 75.Weiner R, Hoffer PB, Thakur ML. Lactoferrin: its role as a Ga-67-binding protein in polymorphonuclear leukocytes. J Nucl Med. 1981;22(1):32–7. [PubMed] [Google Scholar]
  • 76.Kumar V, Boddeti DK. 68Ga-Radiopharmaceuticals for PET Imaging of Infection and Inflammation. In: Baum RP, Rösch F, editors. Theranostics, Gallium-68, and Other Radionuclides: A Pathway to Personalized Diagnosis and Treatment. Berlin, Heidelberg: Springer Berlin Heidelberg; 2013. pp. 189–219. [Google Scholar]
  • 77.Tsan MF. Mechanism of gallium-67 accumulation in inflammatory lesions. J Nucl Med. 1985;26(1):88–92. [PubMed] [Google Scholar]
  • 78.Palestro CJ. The current role of gallium imaging in infection. Semin Nucl Med. 1994;24(2):128–41. doi: 10.1016/s0001-2998(05)80227-2. [DOI] [PubMed] [Google Scholar]
  • 79.Makinen TJ, Lankinen P, Poyhonen T, Jalava J, Aro HT, Roivainen A. Comparison of 18F-FDG and 68Ga PET imaging in the assessment of experimental osteomyelitis due to Staphylococcus aureus. Eur J Nucl Med Mol Imaging. 2005;32(11):1259–68. doi: 10.1007/s00259-005-1841-9. [DOI] [PubMed] [Google Scholar]
  • 80.Kumar V, Boddeti DK, Evans SG, Angelides S. (68)Ga-Citrate-PET for diagnostic imaging of infection in rats and for intra-abdominal infection in a patient. Curr Radiopharm. 2012;5(1):71–5. doi: 10.2174/1874471011205010071. [DOI] [PubMed] [Google Scholar]
  • 81.Nanni C, Errani C, Boriani L, et al. 68Ga-citrate PET/CT for evaluating patients with infections of the bone: preliminary results. Journal of Nuclear Medicine. 2010;51(12):1932–6. doi: 10.2967/jnumed.110.080184. [DOI] [PubMed] [Google Scholar]
  • 82.Vorster M, Maes A, Van de Wiele C, Sathekge M. 68Ga-citrate PET/CT in Tuberculosis: A pilot study. The quarterly journal of nuclear medicine and molecular imaging: official publication of the Italian Association of Nuclear Medicine (AIMN)[and] the International Association of Radiopharmacology (IAR), [and] Section of the Society of. 2014 doi: 10.23736/S1824-4785.16.02680-7. [DOI] [PubMed] [Google Scholar]
  • 83.Kumar V, Boddeti DK, Evans SG, Roesch F, Howman-Giles R. Potential use of 68Ga-apo-transferrin as a PET imaging agent for detecting Staphylococcus aureus infection. Nuclear Medicine and Biology. 2011;38(3):393–8. doi: 10.1016/j.nucmedbio.2010.09.011. [DOI] [PubMed] [Google Scholar]
  • 84.Petrik M, Haas H, Dobrozemsky G, et al. 68Ga-siderophores for PET imaging of invasive pulmonary aspergillosis: proof of principle. J Nucl Med. 2010;51(4):639–45. doi: 10.2967/jnumed.109.072462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ebenhan T, Gheysens O, Kruger HG, Zeevaart JR, Sathekge MM. Antimicrobial peptides: their role as infection-selective tracers for molecular imaging. BioMed research international. 2014;2014 doi: 10.1155/2014/867381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lupetti A, Welling MM, Pauwels EK, Nibbering PH. Radiolabelled antimicrobial peptides for infection detection. The Lancet infectious diseases. 2003;3(4):223–9. doi: 10.1016/s1473-3099(03)00579-6. [DOI] [PubMed] [Google Scholar]
  • 87.Welling MM, Paulusma-Annema A, Balter HS, Pauwels EK, Nibbering PH. Technetium-99m labelled antimicrobial peptides discriminate between bacterial infections and sterile inflammations. European journal of nuclear medicine. 2000;27(3):292–301. doi: 10.1007/s002590050036. [DOI] [PubMed] [Google Scholar]
  • 88.Ferro-Flores G, de Murphy CA, Pedraza-López M, et al. In vitro and in vivo assessment of 99m Tc-UBI specificity for bacteria. Nuclear medicine and biology. 2003;30(6):597–603. doi: 10.1016/s0969-8051(03)00054-4. [DOI] [PubMed] [Google Scholar]
  • 89.Akhtar MS, Qaisar A, Irfanullah J, et al. Antimicrobial peptide 99mTc-ubiquicidin 29–41 as human infection-imaging agent: clinical trial. Journal of Nuclear Medicine. 2005;46(4):567–73. [PubMed] [Google Scholar]
  • 90.Ferro-Flores G, Avila-Rodríguez MA, García-Pérez FO. Imaging of bacteria with radiolabeled ubiquicidin by SPECT and PET techniques. Clinical and Translational Imaging. 2016;4(3):175–82. [Google Scholar]
  • 91.Brouwer CP, Bogaards SJ, Wulferink M, Velders MP, Welling MM. Synthetic peptides derived from human antimicrobial peptide ubiquicidin accumulate at sites of infections and eradicate (multi-drug resistant) Staphylococcus aureus in mice. Peptides. 2006;27(11):2585–91. doi: 10.1016/j.peptides.2006.05.022. [DOI] [PubMed] [Google Scholar]
  • 92.Nibbering PH, Welling MM, Paulusma-Annema A, Brouwer CP, Lupetti A, Pauwels EK. 99mTc-Labeled UBI 29–41 peptide for monitoring the efficacy of antibacterial agents in mice infected with Staphylococcus aureus. Journal of Nuclear Medicine. 2004;45(2):321–6. [PubMed] [Google Scholar]
  • 93.Beiki D, Yousefi G, Fallahi B, et al. 99mtc-Ubiquicidin [29–41], a promising radiopharmaceutical to differentiate orthopedic implant infections from sterile inflammation. Iranian journal of pharmaceutical research: IJPR. 2013;12(2):347. [PMC free article] [PubMed] [Google Scholar]
  • 94.Welling MM, Bunschoten A, Kuil J, et al. Development of a hybrid tracer for SPECT and optical imaging of bacterial infections. Bioconjugate chemistry. 2015;26(5):839–49. doi: 10.1021/acs.bioconjchem.5b00062. [DOI] [PubMed] [Google Scholar]
  • 95.Salber D, Gunawan J, Langen K-J, et al. Comparison of 99mTc-and 18F-Ubiquicidin Autoradiography to Anti–Staphylococcus aureus Immunofluorescence in Rat Muscle Abscesses. Journal of Nuclear Medicine. 2008;49(6):995–9. doi: 10.2967/jnumed.108.050880. [DOI] [PubMed] [Google Scholar]
  • 96.Ebenhan T, Zeevaart JR, Venter JD, et al. Preclinical evaluation of 68Ga-labeled 1, 4, 7-triazacyclononane-1, 4, 7-triacetic acid-ubiquicidin as a radioligand for PET infection imaging. Journal of Nuclear Medicine. 2014;55(2):308–14. doi: 10.2967/jnumed.113.128397. [DOI] [PubMed] [Google Scholar]
  • 97.Vilche M, Reyes AL, Vasilskis E, Oliver P, Balter H, Engler H. 68Ga-NOTA-UBI-29–41 as a PET Tracer for Detection of Bacterial Infection. Journal of Nuclear Medicine. 2016;57(4):622–7. doi: 10.2967/jnumed.115.161265. [DOI] [PubMed] [Google Scholar]
  • 98.Ebenhan T, Sathekge M, Lenagana T, et al. 68Ga-NOTA-functionalized Ubiquicidin: Cytotoxicity, Biodistribution, Radiation Dosimetry and First-in-Human Positron Emission Tomography/Computed Tomography Imaging of Infections. Journal of Nuclear Medicine. 2017 doi: 10.2967/jnumed.117.200048. [DOI] [PubMed] [Google Scholar]
  • 99.Mokaleng BB, Ebenhan T, Ramesh S, et al. Synthesis, 68Ga-radiolabeling, and preliminary in vivo assessment of a depsipeptide-derived compound as a potential PET/CT infection imaging agent. BioMed research international. 2015;2015 doi: 10.1155/2015/284354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ebenhan T, Mokaleng BB, Venter JD, Kruger HG, Zeevaart JR, Sathekge M. Preclinical assessment of a 68Ga-DOTA-functionalized depsipeptide as a radiodiagnostic infection imaging agent. Molecules. 2017;22(9):1403. doi: 10.3390/molecules22091403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Dutta J, Baijnath S, Somboro AM, et al. Synthesis, in vitro evaluation, and 68Ga-radiolabeling of CDP1 toward PET/CT imaging of bacterial infection. Chemical Biology & Drug Design. 2017 doi: 10.1111/cbdd.12980. [DOI] [PubMed] [Google Scholar]
  • 102.Welling MM, Hiemstra PS, van den Barselaar MT, et al. Antibacterial activity of human neutrophil defensins in experimental infections in mice is accompanied by increased leukocyte accumulation. Journal of Clinical Investigation. 1998;102(8):1583. doi: 10.1172/JCI3664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rusckowski M, Qu T, Pullman J, Marcel R. Inflammation and infection imaging with a (99m) Tc-neutrophil elastase inhibitor in monkeys. The Journal of Nuclear Medicine. 2000;41(2):363. [PubMed] [Google Scholar]
  • 104.Liberatore M, Pala A, Scaccianoce S, et al. Microbial Targeting of 99mTc-Labeled Recombinant Human β-Defensin-3 in an Animal Model of Infection: A Feasibility Pilot Study. Journal of Nuclear Medicine. 2009;50(5):823–6. doi: 10.2967/jnumed.108.055533. [DOI] [PubMed] [Google Scholar]
  • 105.Ujula T, Salomäki S, Virsu P, et al. Synthesis, 68 Ga labeling and preliminary evaluation of DOTA peptide binding vascular adhesion protein-1: a potential PET imaging agent for diagnosing osteomyelitis. Nuclear medicine and biology. 2009;36(6):631–41. doi: 10.1016/j.nucmedbio.2009.04.008. [DOI] [PubMed] [Google Scholar]
  • 106.Ordonez AA, Bambarger LE, Jain SK, Weinstein EA. Biodistribution and Pharmacokinetics of Antimicrobials. In: Jain SK, editor. Imaging Infections: From Bench to Bedside. Cham: Springer International Publishing; 2017. pp. 209–22. [Google Scholar]
  • 107.Zhang Z, Gogarty KR, Daryaee F, Tonge PJ. Pharmacokinetic and Pharmacodynamics Relationships. In: Jain SK, editor. Imaging Infections: From Bench to Bedside. Cham: Springer International Publishing; 2017. pp. 195–207. [Google Scholar]
  • 108.DeMarco VP, Ordonez AA, Klunk M, et al. Determination of [11C] rifampin pharmacokinetics within Mycobacterium tuberculosis-infected mice by using dynamic positron emission tomography bioimaging. Antimicrobial agents and chemotherapy. 2015;59(9):5768–74. doi: 10.1128/AAC.01146-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Weinstein E, Liu L, Ordonez A, et al. Noninvasive determination of 2-[18F]-fluoroisonicotinic acid hydrazide pharmacokinetics by positron emission tomography in Mycobacterium tuberculosis-infected mice. Antimicrobial agents and chemotherapy. 2012;56(12):6284–90. doi: 10.1128/AAC.01644-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zhang Z, Ordonez AA, Smith-Jones P, et al. The biodistribution of 5-[18F] fluoropyrazinamide in Mycobacterium tuberculosis-infected mice determined by positron emission tomography. PloS one. 2017;12(2):e0170871. doi: 10.1371/journal.pone.0170871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tsopelas C. Radiotracers used for the scintigraphic detection of infection and inflammation. ScientificWorldJournal. 2015;2015:676719. doi: 10.1155/2015/676719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Dutta J, Naicker T, Ebenhan T, Kruger HG, Arvidsson PI, Govender T. Synthetic approaches to radiochemical probes for imaging of bacterial infections. European Journal of Medicinal Chemistry. 2017 doi: 10.1016/j.ejmech.2017.03.060. [DOI] [PubMed] [Google Scholar]
  • 113.Ebenhan T, Wagener C, Bambarger LE, Kalinda AS, Zeevaart JR. Radiochemistry. In: Jain SK, editor. Imaging Infections: From Bench to Bedside. Cham: Springer International Publishing; 2017. pp. 77–131. [Google Scholar]
  • 114.Auletta S, Galli F, Lauri C, Martinelli D, Santino I, Signore A. Imaging bacteria with radiolabelled quinolones, cephalosporins and siderophores for imaging infection: a systematic review. Clinical and Translational Imaging. 2016;4(4):229–52. doi: 10.1007/s40336-016-0185-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gemmel F, Dumarey N, Welling M. Future diagnostic agents. Semin Nucl Med. 2009;39(1):11–26. doi: 10.1053/j.semnuclmed.2008.08.005. [DOI] [PubMed] [Google Scholar]
  • 116.van Oosten M, Hahn M, Crane LM, et al. Targeted imaging of bacterial infections: advances, hurdles and hopes. FEMS microbiology reviews. 2015;39(6):892–916. doi: 10.1093/femsre/fuv029. [DOI] [PubMed] [Google Scholar]
  • 117.Ordonez AA, Bambarger LE, Murthy N, Wilson DM, Jain SK. Bacterial Imaging. In: Jain SK, editor. Imaging Infections: From Bench to Bedside. Cham: Springer International Publishing; 2017. pp. 149–72. [Google Scholar]
  • 118.Solanki K, Bomanji J, Siraj Q, Small M, Britton K. Tc-99m Infecton: a new class of radiopharmaceutical for imaging infection. Journal of Nuclear Medicine. 1993;34(5):P119-P. [Google Scholar]
  • 119.Britton KE, Vinjamuri S, Hall AV, et al. Clinical evaluation of technetium-99m infecton for the localisation of bacterial infection. European Journal of Nuclear Medicine. 1997;24(5):553–6. doi: 10.1007/BF01267688. [DOI] [PubMed] [Google Scholar]
  • 120.Britton KE, Wareham DW, Das SS, et al. Imaging bacterial infection with 99mTc-ciprofloxacin (Infecton) Journal of Clinical Pathology. 2002;55(11):817–23. doi: 10.1136/jcp.55.11.817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sarda L, Cremieux AC, Lebellec Y, et al. Inability of 99mTc-ciprofloxacin scintigraphy to discriminate between septic and sterile osteoarticular diseases. J Nucl Med. 2003;44(6):920–6. [PubMed] [Google Scholar]
  • 122.Sarda L, Saleh-Mghir A, Peker C, Meulemans A, Cremieux AC, Le Guludec D. Evaluation of (99m)Tc-ciprofloxacin scintigraphy in a rabbit model of Staphylococcus aureus prosthetic joint infection. J Nucl Med. 2002;43(2):239–45. [PubMed] [Google Scholar]
  • 123.Dumarey N, Schoutens A. Renal abscess: filling in with Tc-99m ciprofloxacin of defects seen on Tc-99m DMSA SPECT. Clin Nucl Med. 2003;28(1):68–9. doi: 10.1097/00003072-200301000-00023. [DOI] [PubMed] [Google Scholar]
  • 124.Langer O, Brunner M, Zeitlinger M, et al. In vitro and in vivo evaluation of [18F]ciprofloxacin for the imaging of bacterial infections with PET. Eur J Nucl Med Mol Imaging. 2005;32(2):143–50. doi: 10.1007/s00259-004-1646-2. [DOI] [PubMed] [Google Scholar]
  • 125.Alexander K, Drost WT, Mattoon JS, Kowalski JJ, Funk JA, Crabtree AC. Binding of ciprofloxacin labelled with technetium Tc 99m versus 99mTc-pertechnetate to a live and killed equine isolate of Escherichia coil. Can J Vet Res. 2005;69(4):272–7. [PMC free article] [PubMed] [Google Scholar]
  • 126.Welling M, Stokkel M, Balter J, Sarda-Mantel L, Meulemans A, Le Guludec D. The many roads to infection imaging. Eur J Nucl Med Mol Imaging. 2008;35(4):848–9. doi: 10.1007/s00259-007-0695-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Palestro C, Love C, Caprioli R, et al. Phase II study of 99mTc-ciprofloxacin uptake in patients with high suspicion of osteomyelitis. J NUCL MED MEETING ABSTRACTS. 2006;47(suppl_1):152P. [Google Scholar]
  • 128.Malamitsi J, Papadopoulos A, Vezyrgianni A, Dalianis K, Boutsikou M, Giamarellou H. The value of successive Infecton scans in assessing the presence of chronic bone and joint infection and in predicting its evolution after treatment and after a prolonged follow-up. Nucl Med Commun. 2011;32(11):1060–9. doi: 10.1097/MNM.0b013e32834a837c. [DOI] [PubMed] [Google Scholar]
  • 129.Bhardwaj V, Agrawal M, Suri T, Sural S, Kashyap R, Dhal A. Evaluation of adequacy of short-course chemotherapy for extraspinal osteoarticular tuberculosis using 99mTc ciprofloxacin scan. Int Orthop. 2011;35(12):1869–74. doi: 10.1007/s00264-010-1162-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Tewson T, Yang D, Wong G, et al. The synthesis of fluorine-18 lomefloxacin and its preliminary use in human studies. Nuclear medicine and biology. 1996;23(6):767–72. doi: 10.1016/0969-8051(96)00071-6. [DOI] [PubMed] [Google Scholar]
  • 131.Fischman AJ, Livni E, Babich JW, et al. Pharmacokinetics of [18F] fleroxacin in patients with acute exacerbations of chronic bronchitis and complicated urinary tract infection studied by positron emission tomography. Antimicrobial agents and chemotherapy. 1996;40(3):659–64. doi: 10.1128/aac.40.3.659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Fischman AJ, Babich JW, Alpert NM, et al. Pharmacokinetics of 18F-labeled trovafloxacin in normal and Escherichia coli-infected rats and rabbits studied with positron emission tomography. Clinical Microbiology and Infection. 1997;3(1):63–72. doi: 10.1111/j.1469-0691.1997.tb00253.x. [DOI] [PubMed] [Google Scholar]
  • 133.Mostafa M, Motaleb M, Sakr T. Labeling of ceftriaxone for infective inflammation imaging using 99m Tc eluted from 99 Mo/99m Tc generator based on zirconium molybdate. Applied Radiation and Isotopes. 2010;68(10):1959–63. doi: 10.1016/j.apradiso.2010.04.031. [DOI] [PubMed] [Google Scholar]
  • 134.Fazli A, Salouti M, Mazidi M. 99mTc-ceftriaxone, as a targeting radiopharmaceutical for scintigraphic imaging of infectious foci due to Staphylococcus aureus in mouse model. Journal of Radioanalytical and Nuclear Chemistry. 2013;298(2):1227–33. [Google Scholar]
  • 135.Sohaib M, Khurshid Z, Roohi S. Labelling of ceftriaxone with 99mTc and its bio-evaluation as an infection imaging agent. Journal of Labelled Compounds and Radiopharmaceuticals. 2014;57(11):652–7. doi: 10.1002/jlcr.3235. [DOI] [PubMed] [Google Scholar]
  • 136.Singh B, Babbar A, Sharma S, et al. To evaluate the clinical efficacy of a single vial kit preparation of 99mTc-ceftriaxone (Scintibact) for the diagnosis of orthopedic infections-First results. Journal of Nuclear Medicine. 2010;51(supplement 2):373. [Google Scholar]
  • 137.Kaul A, Hazari PP, Rawat H, et al. Preliminary evaluation of technetium-99m-labeled ceftriaxone: infection imaging agent for the clinical diagnosis of orthopedic infection. International Journal of Infectious Diseases. 2013;17(4):e263–e70. doi: 10.1016/j.ijid.2012.10.011. [DOI] [PubMed] [Google Scholar]
  • 138.Motaleb M, El-Kolaly M, Ibrahim A, El-Bary AA. Study on the preparation and biological evaluation of 99mTc–gatifloxacin and 99mTc–cefepime complexes. Journal of Radioanalytical and Nuclear Chemistry. 2011;289(1):57–65. [Google Scholar]
  • 139.El-Tawoosy M. Preparation and biological distribution of 99mTc-cefazolin complex, a novel agent for detecting sites of infection. Journal of Radioanalytical and Nuclear Chemistry. 2013;298(2):1215–20. [Google Scholar]
  • 140.Motaleb M. Preparation of 99mTc-cefoperazone complex, a novel agent for detecting sites of infection. Journal of Radioanalytical and Nuclear Chemistry. 2007;272(1):167–71. [Google Scholar]
  • 141.Mirshojaei S, Gandomkar M, Najafi R, et al. Radio labeling, quality control and biodistribution of 99mTc-cefotaxime as an infection imaging agent. Journal of Radioanalytical and Nuclear Chemistry. 2011;287(1):21–5. [Google Scholar]
  • 142.Mirshojaei S, Erfani M, Shafiei M. Evaluation of 99mTc-ceftazidime as bacterial infection imaging agent. Journal of Radioanalytical and Nuclear Chemistry. 2013;298(1):19–24. doi: 10.1007/s10967-013-2464-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Gomes BV, Rabiller G, Iglesias F, et al. 99mTc-ceftizoxime scintigraphy in normal rats and abscess induced rats. Revista espanola de medicina nuclear. 2005;24(5):312–8. doi: 10.1157/13079282. [DOI] [PubMed] [Google Scholar]
  • 144.Chattopadhyay S, Ghosh M, Sett S, et al. Preparation and evaluation of 99m Tc-cefuroxime, a potential infection specific imaging agent: A reliable thin layer chromatographic system to delineate impurities from the 99m Tc-antibiotic. Applied Radiation and Isotopes. 2012;70(10):2384–7. doi: 10.1016/j.apradiso.2012.06.007. [DOI] [PubMed] [Google Scholar]
  • 145.Sellmyer MA, Lee I, Hou C, et al. Bacterial infection imaging with [18F]fluoropropyl-trimethoprim. Proceedings of the National Academy of Sciences. 2017;114(31):8372–7. doi: 10.1073/pnas.1703109114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Glaudemans A, Galli F, Pacilio M, Signore A. Leukocyte and bacteria imaging in prosthetic joint infection. Eur Cell Mater. 2013;25:61–77. doi: 10.22203/ecm.v025a05. [DOI] [PubMed] [Google Scholar]
  • 147.Palestro CJ. Seminars in nuclear medicine. Vol. 2015. Elsevier; 2015. Radionuclide imaging of osteomyelitis; pp. 32–46. [DOI] [PubMed] [Google Scholar]
  • 148.Palestro C, Love C, Bhargava K. Labeled leukocyte imaging: current status and future directions. The Quarterly Journal of Nuclear Medicine and Molecular Imaging. 2009;53(1):105. [PubMed] [Google Scholar]
  • 149.Rubin RH, Fischman AJ, Callahan RJ, et al. 111In-labeled nonspecific immunoglobulin scanning in the detection of focal infection. N Engl J Med. 1989;321(14):935–40. doi: 10.1056/NEJM198910053211404. [DOI] [PubMed] [Google Scholar]
  • 150.Horger M, Eschmann SM, Pfannenberg C, et al. The value of SPET/CT in chronic osteomyelitis. European journal of nuclear medicine and molecular imaging. 2003;30(12):1665–73. doi: 10.1007/s00259-003-1321-z. [DOI] [PubMed] [Google Scholar]
  • 151.Graute V, Feist M, Lehner S, et al. Detection of low-grade prosthetic joint infections using 99mTc-antigranulocyte SPECT/CT: initial clinical results. European journal of nuclear medicine and molecular imaging. 2010;37(9):1751–9. doi: 10.1007/s00259-010-1431-3. [DOI] [PubMed] [Google Scholar]
  • 152.Meller J, Ivancevic V, Conrad M, Gratz S. Clinical value of immunoscintigraphy in patients with fever of unknown origin. The Journal of Nuclear Medicine. 1998;39(7):1248. [PubMed] [Google Scholar]
  • 153.Palestro CJ, Glaudemans AW, Dierckx RA. Multiagent imaging of inflammation and infection with radionuclides. Clinical and Translational Imaging. 2013;1(6):385–96. doi: 10.1007/s40336-013-0041-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Palestro CJ. Radionuclide imaging of musculoskeletal infection: a review. Journal of Nuclear Medicine. 2016;57(9):1406–12. doi: 10.2967/jnumed.115.157297. [DOI] [PubMed] [Google Scholar]
  • 155.Wong DW, Dhawan VK, Tanaka T, Mishkin FS, Reese IC, Thadepalli H. Imaging endocarditis with Tc-99m-labeled antibody--an experimental study: concise communication. J Nucl Med. 1982;23(3):229–34. [PubMed] [Google Scholar]
  • 156.Pinkston KL, Singh KV, Gao P, et al. Targeting Pili in Enterococcal Pathogenesis. Infection and Immunity. 2014;82(4):1540–7. doi: 10.1128/IAI.01403-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Rubin RH, Young LS, Hansen WP, et al. Specific and nonspecific imaging of localized Fisher immunotype 1 Pseudomonas aeruginosa infection with radiolabeled monoclonal antibody. J Nucl Med. 1988;29(5):651–6. [PubMed] [Google Scholar]
  • 158.Wiehr S, Warnke P, Rolle A-M, et al. New pathogen-specific immunoPET/MR tracer for molecular imaging of a systemic bacterial infection. Oncotarget. 2016;7(10):10990–1001. doi: 10.18632/oncotarget.7770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Malpani BL, Kadival GV, Samuel AM. Radioimmunoscintigraphic approach for the in vivo detection of tuberculomas--a preliminary study in a rabbit model. Int J Rad Appl Instrum B. 1992;19(1):45–53. doi: 10.1016/0883-2897(92)90184-z. [DOI] [PubMed] [Google Scholar]
  • 160.Lee JD, Shin KH, Cho SN, et al. Immunoscintigraphy in the detection of tuberculosis with radiolabelled antibody fragment against Mycobacterium bovis bacillus Calmette-Guérin: a preliminary study in a rabbit model. European Journal of Nuclear Medicine and Molecular Imaging. 1992;19(12):1011–5. doi: 10.1007/BF00180861. [DOI] [PubMed] [Google Scholar]
  • 161.Oyen WJG, Claessens RAMJ, Boerman OC, van der Meer JWM, Corstens FHM. Specific antibody uptake in tuberculosis? European Journal of Nuclear Medicine. 1993;20(6):568–9. doi: 10.1007/BF00175172. [DOI] [PubMed] [Google Scholar]
  • 162.Gijs M, Aerts A, Impens N, Baatout S, Luxen A. Aptamers as radiopharmaceuticals for nuclear imaging and therapy. Nuclear medicine and biology. 2016;43(4):253–71. doi: 10.1016/j.nucmedbio.2015.09.005. [DOI] [PubMed] [Google Scholar]
  • 163.Sunbul M, Jäschke A. Contact-Mediated Quenching for RNA Imaging in Bacteria with a Fluorophore-Binding Aptamer. Angewandte Chemie International Edition. 2013;52(50):13401–4. doi: 10.1002/anie.201306622. [DOI] [PubMed] [Google Scholar]
  • 164.dos Santos SR, Corrêa CR, de Barros ALB, et al. Identification of Staphylococcus aureus infection by aptamers directly radiolabeled with technetium-99m. Nuclear medicine and biology. 2015;42(3):292–8. doi: 10.1016/j.nucmedbio.2014.12.002. [DOI] [PubMed] [Google Scholar]
  • 165.Ferreira IM, de Sousa Lacerda CM, dos Santos SR, et al. Detection of bacterial infection by a technetium-99m-labeled peptidoglycan aptamer. Biomedicine & Pharmacotherapy. 2017;93:931–8. doi: 10.1016/j.biopha.2017.07.017. [DOI] [PubMed] [Google Scholar]
  • 166.Chen L, Wang Y, Cheng D, et al. 99m Tc-MORF oligomers specific for bacterial ribosomal RNA as potential specific infection imaging agents. Bioorganic & medicinal chemistry. 2013;21(21):6523–30. doi: 10.1016/j.bmc.2013.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Lazzeri E. Systematic review of in vivo microorganisms imaging with labeled vitamins, bacteriophages and oligomers. Clinical and Translational Imaging. 2016;4(4):265–72. [Google Scholar]
  • 168.Drulis-Kawa Z, Majkowska-Skrobek G, Maciejewska B, Delattre A-S, Lavigne R. Learning from bacteriophages-advantages and limitations of phage and phage-encoded protein applications. Current Protein and Peptide Science. 2012;13(8):699–722. doi: 10.2174/138920312804871193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Rusckowski M, Gupta S, Liu G, Dou S, Hnatowich DJ. Investigations of a 99mTc-labeled bacteriophage as a potential infection-specific imaging agent. Journal of Nuclear Medicine. 2004;45(7):1201–8. [PubMed] [Google Scholar]
  • 170.Rusckowski M, Gupta S, Liu G, Dou S, Hnatowich DJ. Investigation of four 99m Tc-labeled bacteriophages for infection-specific imaging. Nuclear medicine and biology. 2008;35(4):433–40. doi: 10.1016/j.nucmedbio.2008.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Cardoso ME, Fernández L, Tejería E, Esperón P, Terán M. Evaluation of a labelled bacteriophage with 99mTc as a potential agent for infection diagnosis. Current Radiopharmaceuticals. 2016;9(2):137–42. doi: 10.2174/1874471009666160215162023. [DOI] [PubMed] [Google Scholar]
  • 172.Kennedy DO. B Vitamins and the Brain: Mechanisms, Dose and Efficacy--A Review. Nutrients. 2016;8(2):68. doi: 10.3390/nu8020068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Rusckowski M, Fritz B, Hnatowich DJ. Localization of infection using streptavidin and biotin: an alternative to nonspecific polyclonal immunoglobulin. J Nucl Med. 1992;33(10):1810–5. [PubMed] [Google Scholar]
  • 174.Lazzeri E, Erba P, Perri M, Doria R, Tascini C, Mariani G. Clinical impact of SPECT/CT with In-111 biotin on the management of patients with suspected spine infection. Clin Nucl Med. 2010;35(1):12–7. doi: 10.1097/RLU.0b013e3181c36173. [DOI] [PubMed] [Google Scholar]
  • 175.Lazzeri E, Erba P, Perri M, et al. Scintigraphic imaging of vertebral osteomyelitis with 111in-biotin. Spine (Phila Pa 1976) 2008;33(7):E198–204. doi: 10.1097/BRS.0b013e31816960c9. [DOI] [PubMed] [Google Scholar]
  • 176.Lazzeri E, Pauwels EK, Erba PA, et al. Clinical feasibility of two-step streptavidin/111In-biotin scintigraphy in patients with suspected vertebral osteomyelitis. European journal of nuclear medicine and molecular imaging. 2004;31(11):1505–11. doi: 10.1007/s00259-004-1581-2. [DOI] [PubMed] [Google Scholar]
  • 177.Shoup TM, Fischman AJ, Jaywook S, Babich JW, Strauss HW, Elmaleh DR. Synthesis of fluorine-18-labeled biotin derivatives: biodistribution and infection localization. J Nucl Med. 1994;35(10):1685–90. [PubMed] [Google Scholar]
  • 178.Baldoni D, Waibel R, Bläuenstein P, et al. Evaluation of a Novel Tc-99m Labelled Vitamin B12 Derivative for Targeting Escherichia coli and Staphylococcus aureus In Vitro and in an Experimental Foreign-Body Infection Model. Molecular Imaging and Biology. 2015;17(6):829–37. doi: 10.1007/s11307-015-0832-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Leevy WM, Gammon ST, Jiang H, et al. Optical imaging of bacterial infection in living mice using a fluorescent near-infrared molecular probe. J Am Chem Soc. 2006;128(51):16476–7. doi: 10.1021/ja0665592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Leevy WM, Gammon ST, Johnson JR, et al. Noninvasive optical imaging of staphylococcus aureus bacterial infection in living mice using a Bis-dipicolylamine-Zinc(II) affinity group conjugated to a near-infrared fluorophore. Bioconjug Chem. 2008;19(3):686–92. doi: 10.1021/bc700376v. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Liu X, Cheng D, Gray BD, et al. Radiolabeled Zn-DPA as a potential infection imaging agent. Nucl Med Biol. 2012;39(5):709–14. doi: 10.1016/j.nucmedbio.2011.12.006. [DOI] [PubMed] [Google Scholar]
  • 182.Rice DR, Plaunt AJ, Turkyilmaz S, et al. Evaluation of [(1)(1)(1)In]-labeled zinc-dipicolylamine tracers for SPECT imaging of bacterial infection. Mol Imaging Biol. 2015;17(2):204–13. doi: 10.1007/s11307-014-0758-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Rice DR, Clear KJ, Smith BD. Imaging and therapeutic applications of zinc (II)-dipicolylamine molecular probes for anionic biomembranes. Chemical Communications. 2016;52(57):8787–801. doi: 10.1039/c6cc03669d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wyffels L, Gray BD, Barber C, et al. Synthesis and preliminary evaluation of radiolabeled bis(zinc(II)-dipicolylamine) coordination complexes as cell death imaging agents. Bioorg Med Chem. 2011;19(11):3425–33. doi: 10.1016/j.bmc.2011.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Wang H, Tang X, Tang G, et al. Noninvasive positron emission tomography imaging of cell death using a novel small-molecule probe, (18)F labeled bis(zinc(II)-dipicolylamine) complex. Apoptosis. 2013;18(8):1017–27. doi: 10.1007/s10495-013-0852-4. [DOI] [PubMed] [Google Scholar]
  • 186.Sun T, Tang G, Tian H, et al. Positron emission tomography imaging of cardiomyocyte apoptosis with a novel molecule probe [18F]FP-DPAZn2. Oncotarget. 2015;6(31):30579–91. doi: 10.18632/oncotarget.5769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Panizzi P, Nahrendorf M, Figueiredo JL, et al. In vivo detection of Staphylococcus aureus endocarditis by targeting pathogen-specific prothrombin activation. Nat Med. 2011;17(9):1142–6. doi: 10.1038/nm.2423. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES