Abstract
Infection is ubiquitous. However, its management is challenging for both the patients and the healthcare providers. Scintigraphic imaging of infection dates back nearly half a century. The advances in our understanding of the pathophysiology of disease at cellular and molecular levels have paved the way to the development of a large number of radiopharmaceuticals for scintigraphic imaging of infection. These include radiolabeling of blood elements such as serum proteins, white blood cells (WBCs), and cytokines, to name a few. Infectious foci have also been imaged using a radiolabeled sugar molecule by taking advantage of increased metabolic activity in the infectious lesions. Literature over the years has well documented that none of the radiopharmaceuticals and associated procedures that facilitate imaging infection are flawless and acceptable without a compromise. As a result, only a few compounds such as 99mTc-hexamethylpropyleneamineoxime, 18F-FDG, the oldest but still considered as a gold standard 111In-oxine, and, yes, even 67Ga-citrate in some countries, have remained in routine clinical practice. Nonetheless, the interest of scientists and physicians to improve the approaches to imaging and to the management of infection is noteworthy. These approaches have paved the way for the development of numerous, innovative radiopharmaceuticals to label autologous WBCs ex vivo or even those that could be injected directly to image infection or inflammation without direct involvement of WBCs. In this review, we briefly describe these agents with their pros and cons and place them together for future reference.
Introduction
Infection is a major problem for patients who experience it and clinicians who manage the disease. Accurate and early diagnosis can be difficult and time-consuming, whereas delays in diagnosis can be life-threatening. Therefore, accurate detection and localization of infection and inflammation at an early stage is of vital importance for patient management, as well as for the cost containment. History, physical examination, various laboratory tests such as erythrocyte sedimentation rate, and C-reactive protein measurement are performed to determine onset of infection. For localization of infection, however, radiologic methods such as X-ray, ultrasonography, computed tomography, magnetic resonance imaging, and nuclear medicine imaging methods are used. These methods, however, suffer from limitations and cannot reliably detect infection at an early stage.
Most radiopharmaceuticals required for nuclear medicine imaging are designed to accumulate in infection by increased capillary blood flow and increased vascular permeability, or are associated with migration of leukocytes. In general, therefore, nuclear medicine imaging of infection is derived from the pathophysiologic course of infection, and can detect infection and inflammation in an early phase before the appearance of morphologic changes at the site of infection.1–6
The quest for scintigraphic imaging of infection dates back to early 1970s, following the serendipitous observation of accumulation of 67Gallium (67Ga) citrate, 4 days following its administration for imaging Hodgkin disease.7 However, 1976 marked a new chapter in the history of imaging infection; McAfee and Thakur surveyed nearly 100 radioactive compounds for labeling autologous white blood cells (WBCs) ex vivo.8,9 The survey led to the development of 111Indium (111In)-oxine as the most efficient liqid-soluble agent, to label WBCs ex vivo and to image experimental abscesses in an animal model10 and pyogenic abscesses in man.11 Although now more than 4 decades have elapsed, 111In-oxine is still considered as gold standard for WBC labeling in routine practice for imaging infection. Nonetheless, the technique of labeling WBCs ex vivo suffers from numerous limitations and falls short of the ideal requirements outlined in the Table.4,5 To address these issues over the years, a score of novel radioactive compounds with innovative hypotheses have been evaluated.1–4
Table.
– Specific to infection |
– Selective accumulation in infection foci |
– No uptake in noninflamed sites |
– No side effects, no toxicity, no immunogenic response |
– Fast clearance from normal tissues |
– Low cost |
– Easy preparation |
– The ability to distinguish infection from inflammation |
– Applicable for use in immunocompromised patients |
– Low marrow and renal accumulation |
This review briefly describes these agents, including their strengths and limitations,
Radiopharmaceuticals
67Ga-Citrate
67Ga-citrate was one of the first radiopharmaceuticals used for scintigraphic imaging of infection.7 67Ga-citrate, after intravenous (i.v.) administration, binds primarily to transferrin and to other iron binding proteins such as lactoferrin, ferritin, and bacterial siderophores. A large percentage is excreted via the kidneys in the first 24 hours after injection and relatively small proportion in bowel in the first week. Radioactivity therefore is seen in the kidneys, bladder, abdominal region, and in organs rich in lactoferrin such as the eyes and lactating breasts. Physiologic activity is also seen in the bone, bone marrow, liver, spleen, and soft tissues. Forty-eight hours after its administration, variable physiologic activity is also seen in the lacrimal glands, salivary glands, and breast tissue (Fig). In children, uptake in the thymus is also observed.
Figure.
Biodistribution in man of 14 most commonly used radiopharmaceuticals injected intravenously. 67Ga-citrate and Fluorine-18-FDG are injected directly and 111In (oxine) and 99mTc (HMPAO) WBC are injected after they are labeled ex vivo. Although different organ distribution of 67Ga-citrate and 18F-FDG should be expected, the distribution of 111In-WBC and 99mTc-WBC should be similar. However it is not. Rapid in vivo, release of 99mTc from labeled WBC and its subsequent distribution different from those of the WBC, contribute to the difference and may lead to false-positive or false-negative images.
67Ga-citrate is sensitive for imaging acute infection, chronic infection, and inflammation. 67Ga-citrate imaging is also useful in fever of unknown origin (FUO), vertebral osteomyelitis, and opportunistic infections in immunocompromised patients. However, its specificity is compromised because of uptake in many normal tissues as well as in tumors. A major drawback of 67Ga-citrate, however, is its slow blood clearance, which compels imaging to be performed at 48 and sometimes up to 72–96 hours after administration.2,4,6,12
Ex Vivo Labeling of Leukocytes
111In-Oxine
With the hypothesis that the above lack of specificity of 67Ga-citrate could be eliminated with the use of autologous WBCs, which spontaneously migrate to foci of infection physiologically, McAfee and Thakur initiated their survey of agents to radiolabel WBC for imaging infection.8,9 Subsequently, conclusion that 111In-oxine was the agent of choice to label WBC for imaging infection was drawn.10,11
For such labeling, venous blood is drawn from the patient into a sterile, disposable plastic syringe. Red cells are sedimented, and the carefully separated leukocyte-rich plasma is centrifuged to obtain WBCs. After these cells are labeled with 111In, they are reinfused back into the patient intravenously.1
Thakur et al11 also demonstrated that 111In-oxine, a lipid-soluble agent, passively diffuses through the cell membrane and binds to cytoplasmic components of the cells, provides a stable label, and serves to follow the cell trafficking in vivo. 111In-labeled leukocytes once injected intravenously accumulate in the lungs in a large proportion and then clear from the lungs in approximately 4 hours. 111In-labeled leukocytes also accumulate in the spleen, liver, and bone marrow. Accumulation of radioactivity in the spleen and liver is also supplemented by red cells, lymphocytes, and platelets that may be present in the separated autologous leukocytes and labeled with 111In. Although patient imaging can be performed 4–6 hours post injection of labeled leukocytes, best results are achieved at imaging 24 hours later.1,11,12
Radiolabeled WBCs are used to image FUO, osteomyelitis, infected orthopedic hardware, inflammatory bowel disease (IBD), abdominal infection, pelvic sepsis, and other inflammatory processes. Sensitivity of this method is ~90% for acute and chronic infection. In vitro WBC labeling involves multiple steps and is time-consuming. In addition, there is a possible risk associated with ex vivo handling of blood that may be tainted with hepatitis B, hepatitis C, or HIV.1,2
Other 111In Agents
111In-oxine binds to transferrin in plasma and requires plasma to be removed during cell labeling. This may compromise cell viability. To address this issue, lipophilic agents such as 111In-acetylacetone (ACAC) and tropolone were developed.11,13,14
In contrast to 111In-oxine, 111In-ACAC and 111In-tropolone have been shown to label WBC in a plasma medium. 111In-tropolone-labeled WBC clear from the lungs in a shorter period of time than those WBC labeled with 111In-oxine in saline. In addition, 111In-tropolone-labeled WBC migrate to the inflammation quickly, therefore infections can be detected in less than 4 hours after injection. However, some investigators observed that 111In-tropolone-labeled WBC had compromised chemotactic characteristics. Other authors found 111In-ACAC and 111In-tropolone were less appealing than 111In-oxine to label WBC for imaging infection.13,15
In 1985, simple kit formulation of 111 In-2-mercaptopyridine-N-oxide (Merc), a nontoxic, water-soluble agent was evaluated to label human platelets efficiently. This new agent was also evaluated for labeling canine leukocytes in plasma.13 MERC binds 111In as avidly as tropolone, provides labeling in a plasma medium, and does not affect cell viability. 111In-Merc-labeled leukocytes are cleared from the lungs quickly and accumulate in the abscesses rapidly.13–15 The preparation process for labeling WBC with 111In-Merc was also less cumbersome than 111In-oxine. However, 111In-Merc did not become a widely used agent for WBC labeling.2,7,15
99mTc HMPAO
99mTc-hexamethylpropyleneamineoxime (HMPAO) is a second-generation radiopharmaceutical to label leukocytes introduced by Peters et al in 1986.16,17 99mTc-HMPAO is a lipophilic agent that labels WBC predominantly. HMPAO is available commercially as a lyophilized powder (kit) ready to be labeled when needed with generator-produced 99mTc. Alternatively, 99mTc-HMPAO can be bought commercially as a single-use dose. The half-life of 99mTc (t1/2: 6 hours) is long enough to allow imaging to be carried out without excessive radioactivity decay having occurred, but not too long to induce unnecessary radiation burden long after imaging has been performed. In addition to the lower radiation dose of 99mTc than 111In, the decay characteristics of 99mTc (140 Kev) are more favorable to image using a gamma camera. Image quality with 99mTc is therefore better than with 111In. Because of the low radiation dose with 99mTc-WBC, imaging can be repeated if required.18,19 99mTc elutes out of the WBCs labeled with 99mTc-HMPAO. This, in part, leads to a very different distribution of 99mTc-HMPAO-WBC than 111In-oxine-labeled WBC. With 99mTc-HMPAO-labeled WBC, radioactivity is seen in urine, bladder, gall bladder, bone marrow, and gastrointestinal system, especially in the colon. Nearly 6% of 99mTc is excreted in the stool in 48 hours.1,2,16 Also, nonspecific activity is seen in the kidneys. Such a distribution of radioactivity contributes to false-positive results. 99mTc-WBC is therefore less useful for imaging abdominal infection. For imaging renal, genitourinary, and gastrointestinal infection, therefore, 111In-WBC is preferred.1,12,20
18F-FDG
Over the past few years, 18F-FDG has been increasingly used for imaging infection. The use of 18F-FDG is driven by its 2 important virtues. First, 18F is a cyclotron-produced positron emitting radionuclide that facilitates PET imaging and the other is that 18F-FDG is taken up by WBC through glucose transporter, GLUT-1, and then is phosphorylated, allowing 18F to monitor WBC distribution in vivo. For imaging infection, 18F-FDG is used in 2 ways: (1) by labeling WBC ex vivo and (2) by injecting it directly in vivo.21–23
Ex Vivo 18F-FDG-Labeled WBC
Rini et al24 in 2006, labeled WBC ex vivo and PET-imaged infection in vivo. This investigation provided promising results. However, the short half-life of 18F (t1/2-110 minutes) is practically too short for the relatively lengthy ex vivo WBC labeling procedure, and prevents delayed imaging when needed. Furthermore, in various studies, it was reported that the WBC labeling efficiency (LE) with 18F-FDG was much lower and more variable than with 111In oxine. Although not examined in detail, the leukocyte glucose transporter expression, the serum glucose levels, and the presence of intrinsic proteins have been thought to affect the LE. Additionally, excessive wash-out of 18F from 18F-FDG-labeled WBC was also discouraging. These prevented the use of 18F-FDG-WBC from routine clinical practice from imaging infection.25,26
Directly Injected 18F-FDG
Directly injected 18F-FDG has therefore remained a much preferable approach to image infection. Not only is the approach convenient but it also provides high sensitivity for imaging infection. The limitations, however, are the lack of specificity rendered by excessive 18F-FDG uptake by the metabolic activity of many normal organs such as the heart, brain, and muscle, as well as in the kidneys and bladder caused by urinary excretion. 18F-FDG for imaging infection requires the patient to fast before injection and in the case of diabetic patients, blood glucose levels to be monitored before the administration of 18F-FDG.27–30
18F-FDG-PET/CT is used for evaluation of patients with FUO with high sensitivity (90%) and specificity (97%). 18F-FDG PET/CT has a high negative predictive value for determining etiologies of FUO (negative predictive value 100%). Furthermore, 18F-FDG-PET/CT imaging has high diagnostic value for the evaluation of children with FUO and unexplained origin of inflammation.31–33
Other PET Imaging Compounds
64Cu-Tropolone
Copper-64 (64Cu) is an emerging positron emitting radionuclide used extensively primarily in preclinical and translational oncologic PET imaging. Chelated with tropolone 64Cu was investigated to label WBC.25 Poor intra-WBC retention of 64Cu prompted Bhargava and colleagues26,34 to add to the WBC incubation mixture a membrane-permeable fluorinated divalent chelator MF/AM (2-(2-amino-4-methyl-5fl ophenoxy) methyl-8-aminoquinoline-N, N, N’, N’-tetra-acetic acid), which improved labeling yield as well as the 64Cu-intra-WBC retention. Post-labeled cell viability was not compromised. Although no further studies could be found, the longer half-life of 64Cu may be a better substitute for PET imaging of infection than 18F-FDG-WBC.
89Zr Compounds
89Zirconium (89Zr) (t1/2: 78.4 hours, β+ 22%) compounds for labeling WBCs have been also investigated. In 1 investigation, 89Zr-or 64Cu-labeled chitosan (CN) nanoparticles were used to label WBC presumably by phagocytosis. Mixed WBCs were separated from human blood. 89Zr-CN-labeled WBCs had higher LE than 64Cu-CN-labeled WBCs.35 89Zr-CN-labeled WBCs showed less elution of radioactivity than 64Cu-CN-labeled WBC. Although the 78.4-hour half-life of 89Zr could be considered too long for PET imaging of infection, the 89Zr tracer shall be highly useful to follow the in vivo kinetics of such cells as T cells or platelets.
To follow in vivo cell trafficking, Sato et al developed a 89Zr-oxine complex for labeling dendritic cells, cytotoxic T cells, and natural killer cells.36 Labeling did not compromise the viability of dendritic cells and cytotoxic T cells, and function of T cells. The lipid solubility of 89Zr-oxine made it useful to label.
Another compound of 89Zr namely 89Zr-oxalate was also investigated to image inflammation and tumors. Although exact mechanism of inflammatory cell binding was not determined, tumor imaging results showed that 89Zr-oxalate had higher uptake in macrophages than other cells in the tumor. The authors also reported that 89Zr-oxalate had higher tumor specificity than 18F-FDG. Data suggest that exploring the use of 89Zr-oxalate to label WBCs ex vivo or even in vivo may be worthwhile.37
In Vivo Labeling of WBC
The advent of receptor-specific monoclonal antibodies (mAb) combined with the better understanding of the receptor expression on human WBC led to the field of labeling human WBC in vivo using radiolabeled mAbs. The major thrust of these investigations was driven by the objectives of eliminating the disadvantages associated with the procedures required to label WBC ex vivo. Although several mAbs were evaluated, 4 were examined in humans. Three of them targeted various epitopes of nonspecific cross-reacting antigen (NCA), namely NCA-47, NCA-90, and NCA-97. The fourth one targeted stage-specific embryonic antigen-1 (SSEA-1). All of them were of murine origin and were not humanized. The pros and cons of them are briefly described below.
NCA-47 Antibody
The anti NCA-47 was the first murine mAb (IgG1) that was labeled with 123Iodine (123I) (t1/2: 13.3 hours), 159 KeV (83%), and investigated to image infection in humans. All inflammatory or infectious lesions were successfully imaged without adverse events.38,39
Anti NCA-90
This anti NCA-90 antibody presented 2 advantages over the anti NCA-47 antibody stated above. First, the antibody (IMMU-MN3) was fragmented (Fab’), which facilitated faster clearance; and second it was labeled with a generator-produced and commonly used radionuclide 99mTc (t1/2: 6.1 hours, γ−140 KeV: 90%). Also known as LeukoScan, the mAb is commercially available in Europe and has been used extensively in humans. After i.v. administration, approximately 4% of administered 99mTc was bound to circulating neutrophils. In vivo elution of 99mTc was reported. Despite the limitations, the convenience makes its use attractive. It is used in humans to image osteomyelitis, prosthetic joint infections, soft tissue infections, and pulmonary aspergillosis. False-positive results in patients were reported.40,41
Anti NCA-95 (99mTc-BW250/183)
A murine monoclonal IgG1 antibody, BW250/183 (Behring-Werke, Marburg), also binds to NCA-95 receptors on the neutrophils.42 It was labeled with 99mTc. At least 10% of this agent was bound to granulocytes in blood at 45 minutes after i.v. administration. An important and serious side effect of 99mTc-labeled BW250/183 (besilesomab, SantimumR) is dose-dependent human anti-mouse antibody formation. Uptake of 99mTc-BW250/183 was very high in the spleen and bone marrow, whereas the uptake in infection foci was low. Moreover, higher dose of 99mTc-BW250/183 caused neutropenia that took several hours for recovery.43,44
99mTc-Anti-SSEA-1 (Anti Cluster Differentiation-15)
Anti-SSEA-1 is an IgM class of mAb and it is produced by immunizing mice with murine embryonal carcinoma F9 cells. SSEA-1 antigen cluster differentiation-15 is expressed on human granulocytes.45 Thakur and colleagues labeled murine IgM monoclonal anti-SSEA-1 (MCA-480) with 99mTc, also called 99mTc-fanolesomab, LeuTech, or NeutroSpec. It binds to CD15 glycoproteins with high affinity (10−11). CD15 is expressed on the human granulocyte membranes in high density. The authors found that anti-SSEA-1 was the most desirable agent for specific labeling human neutrophil in vivo, as approximately 50% of the injected 99mTc was bound to circulating polymorphonuclear leukocytes (PMNs) quickly after i.v. injection.45–48
It was shown that 99mTc-fanolesomab played an important role in imaging equivocal appendicitis. Sensitivity, specificity, and accuracy were found to be 91%, 86%, 87%, respectively. The sensitivity, specificity, and accuracy for imaging osteomyelitis were 90%, 67%, 76%, respectively.49 This agent was investigated on patients with clinical evidence of acute inflammatory processes at the time of imaging. All these patients had accurate positive images.46 99mTc-fanolesomab also imaged prosthetic vascular graft infection safely and accurately (95%). No adverse reactions were reported in patients by any user.50 It was approved by the US Food and Drug Administration for suspected appendicitis in 2004. Later, however, it caused serious adverse reactions in 2 patients, which resulted in its withdrawal from market in late 2005.51–53
Other Monoclonal Antibodies
Radiolabeled Anti-Tumor Necrosis Factor-Alpha Monoclonal Antibody
Tumor necrosis factor-alpha (TNFα) is a proinflammatory cytokine that plays an important role in inflammation.54 Anti-TNF antibodies such as infliximab, adalimumab, golimumab, certolizumab pegol, and etanercept have been developed to treat inflammatory diseases such as rheumatoid arthritis (RA), spondyloarthropathy, psoriatic arthritis, and IBDs.55–61
Among them, infliximab was labeled with 99mTc and evaluated to image persistent knee arthritis. Scintigraphy showed high uptake in the affected knee, which had a high level of TNF before the treatment. Four months after treatment, there was no uptake in the affected knee, so scintigraphy verified the efficiency of the treatment.62
99mTc-infliximab scintigraphy was also used in patients with active and refractory monoarthritis. Target-to-background ratios were higher in patients with affected joints than nonaffected joints before treatment. The authors concluded that 99mTc-anti-TNF mAb scintigraphy can play an important role for choosing the best treatment option for each patient and can minimize the number of unnecessary anti-TNF treatments.63
Adalimumab labeled with 99mTc using succinimidyl-hydrazinonicotinamide (S-HYNIC) was used for imaging patients with RA.64 In this study, inflamed joints were distinctly visualized, but the uptake was not prominent in smaller inflamed joints, probably because of the absence of TNF in those joints.65
Malviya et al evaluated both 99mTc-labeled infliximab and adalimumab for scintigraphic imaging of RA in patients, before and 3 months after intra-articular treatment with infliximab or systemic therapy with adalimumab. After the treatment, 99mTc-anti-TNF mAb scintigraphic images showed decreased uptake in the affected joints, which correlated with the improved clinical symptoms.66
Radiolabeled Anti-CD4 Monoclonal Antibody
CD4 antigen is located on T-cells and macrophages. CD4 plays an important role in RA. CD4-specific scintigraphy was therefore thought to be a useful method for imaging patients with RA, to improve diagnosis and treatment of the disease. 99mTc-labeled anti-human CD4 mAb (MAX. 16H5) showed increased uptake in the RA lesions.67,68
Steinhoff et al69 performed 99mTc-anti-CD4-Fab (99mTc-EP1645) scintigraphy for evaluation of inflammatory activity in patients with RA. They also assessed the safety and tolerability of 99mTc-anti-CD4-Fab (99mTc-EP1645) in patients with active synovitis secondary to RA and determined the potential of 99mTc-anti-CD4-Fab (99mTc-EP1645) as a marker to assess the disease activity in a phase I study. The results demonstrated that 99mTc-anti-CD4-Fab (99mTc-EP1645) scintigraphy was a promising approach to evaluate CD4-specific inflammatory activity and to determine the effectiveness of RA therapy.
Radiolabeled Anti-CD20 Monoclonal Antibody
CD20 is expressed on the surface of both normal and malign T lymphocytes. Rituximab is a chimeric murine or human mAb against the CD20 antigen, and has been used for treatment of active RA resistant to TNF.
Rituximab labeled with 99mTc was used in patients to image various inflammatory and autoimmune diseases including RA. Significant uptake was observed in the inflamed joints, and it was correlated with clinical findings of patients. No adverse events were noted.70,71
Radiolabeled Anti-CD3 Monoclonal Antibody
CD3 is expressed on the T lymphocytes. The murine mAb, muromonab, namely OKT-3, is specific for the CD3 receptor. Muromonab is an immunosuppressant used to prevent acute rejection in transplant patients.
99mTc-labeled anti-CD3 mAb, muromonab (OKT-3) was used in patients with RA to show inflamed synovium.72 99mTc-OKT-3 uptake was seen in painful joints. 99mTc-anti-CD3 mAb imaging can be preferred for assessing the treatment effectiveness in patients with RA. However, utilization of it was abandoned because of the side effects.73,74
A humanized anti-CD3 mAb, visilizumab (Nuvion), was also labeled with 99mTc and used in animals. But no further work in human was noted.75
Anti-E-Selectin Monoclonal Antibody
selectin, a 115 kDa glycoprotein, and endothelial-specific adhesion molecule, is expressed on the luminal surface of the vascular endothelium. The expression of vascular endothelial E-selectin, which plays an important role in the inflammation, is stimulated by proinflammatory cytokines, and promotes adhesion of neutrophils, monocytes, and eosinophils to activated vascular endothelium through carbohydrate ligands such as sialyl Lewis X. The increased expression of E-selectin occurs in various inflammatory diseases, such as RA and IBD. 111In-labeled anti-E-selectin mAb imaging was successfully performed initially in an animal model and then also in humans.76–79
Bhatti et al showed that 111In-labeled anti-E-selectin mAb imaging can localize affected parts of bowel, the extent of Crohn’s disease, and ulcerative colitis. 111In-labeled anti-E-selectin mAb imaging was reported to be superior to 99mTc leukocyte imaging.76
Jamar et al77,78 compared 99mTc-human immunoglobulin (HIG) and a 111In-anti-E-selectin mAb (1.2B6) in patients with RA and synovitis. The investigators noted that 111In-labeled anti-E-selectin mAb was more sensitive, and specific than 99mTc-HIG scintigraphy. 99mTc-labeled anti-E-selectin (1.2B6) Fab fragment was more specific than 99mTc-hydroxydiphosphonate bone scintigraphy shows synovitis in another study.78,79
Human Polyclonal Immunoglobulin-G
HIG labeled with 99mTc or 111In has also been used to image infection or inflammation. Three possible mechanisms of accumulation, namely, capillary leak, binding Fc part of IgG to Fc receptors on granulocytes, and binding of IgG to bacteria are postulated. Radiolabeled HIG has been shown to be suitable for FUO in particular immunodeficient patients. The disadvantage, however, is its high blood pool activity that compromises its usefulness in routine clinical practice.80,81 99mTc-HIG is able to show actively inflamed joints, even in patients with serum-negative RA, in a greater extent than other anatomical imaging methods. Sensitivity and specificity of 99mTc-HIG are reported to be 83.3% and 92%, respectively.82
Radiolabeled Antibiotics
In an attempt to differentiate bacterial infection and sterile inflammation, antibiotics that penetrate bacterial cell wall and kill microorganisms by various mechanisms were radiolabeled and evaluated.83,84
The most commonly used among them to image infection is 99mTc-ciprofloxacin (Infecton).85 Ciprofloxacin is a synthetic broad-spectrum fluoroquinolone antibiotic that binds to bacterial DNA gyrase and inhibits DNA synthesis.86,87 99mTc-ciprofloxacin is primarily excreted from the kidneys and has low liver and bowel uptake.88 Ciprofloxacin was also labeled with 68Ga and 18F.89,90 68Ga-ciprofloxacin was shown to be a good bacteria-specific imaging agent in an Staphylococcus aureus (S. aureus) infected rat model. 18F-ciprofloxacin, however, was not a successful imaging agent.89–91
Clinical studies of 99mTc-Infecton have concluded that Infecton is a good bacterial infection imaging agent for the assessment of tuberculosis, FUO, osteomyelitis, orthopedic prosthesis, spinal infection, abdominal infections, and infection in immunosuppressed patients.92–95 Less promising results, however, have been reported by other investigators.96,97
Other promising antibiotics have been 99mTc-cefazolin, 99mTc-cefepime, 99mTc-clarithromycin, 99mTc-rufloxacin, 99mTc-ceftriaxone, 99mTc-levofloxacin, 99mTc-gemifloxacin, and 99mTc-sitafloxacin, with target-to-nontarget ratio higher than 8.84
Additionally, radiolabeled antibiotics such as 99mTc-rifampicin, 99mTc-nitrofurantoin, 99mTc-amoxicillin, 99mTc-alafosfalin, 2-deoxy-2-[18F]fluoroacetamido-D-glucopyranose, 99mTc-vancomycin, 99mTc-kanamycin, 99mTc-erythromycin, 99mTc-azithromycin, 99mTc-clarithromycin, 99mTc-clindamycin, 99mTc-mebendazole, and 99mTc-2,2'-[(8-hydroxyquinolin-7-yl) methylazanediyl] diacetic acid complex have also evaluated.98–110
Fungal Imaging
Radiolabeled Fluconazole
Among the triazole antifungal agents, fluconazole is the most commonly used compound for the treatment of Candida infections in severely immunocompromised patients. Fluconazole inhibits ergosterol synthesis with binding to cytochrome P450. 99mTc-labeled fluconazole is a promising imaging agent for imaging Candida albicans infections. 99mTc-fluconazole is quickly excreted via the kidneys and has low liver uptake. However, 99mTc-fluconazole accumulated poorly in bacterial infections and in Aspergillus fumigatus infections in mice. Fluconazole was also labeled with 18F for PET imaging. 18F-labeled fluconazole is more lipophilic than 99mTc-labeled fluconazole, so it showed higher accumulation in liver than 99mTc-labeled fluconazole. Uptake of 18F-fluconazole in infection foci was also low.111,112
Radiolabeled Chitinase
Chitin is found in fungal cell and renders chitin a promising candidate to image fungal infection.113,114 123I-labeled chitinase B was investigated in fungal infection in mice. Results were encouraging.114 Chitin binding protein (CBP21) was also labeled with 99mTc via a chelating agent HYNIC. Uptake of this agent in fungal infections was higher than in bacterial infections. The authors concluded that it is a new promising agent to distinguish fungal from bacterial infection.113
Nanoparticles
Two types of nanoparticles namely human serum albumin nanocolloids and lipid liposomes, a few nanometer in size, were labeled with either 99mTc or 111In, and injected directly to image infection in experimental animals and in humans.115–122 The sensitivity of imaging infection (87%) in 1 study was as good as or better than 111In-labeled leukocyte scintigraphy (81%).122 Regional “spilling” of the tracer into the extravascular space through increased capillary permeability was proposed as the probable mechanism of uptake.123 The wide-spread use of these nanoparticles in routine clinical practice was compromised probably because of the adverse reactions the use of particles presented.124
Vitamins
Biotin
Biotin (vitamin H) binds to avidin, a protein commonly available in the eggs of avians, amphibians, and reptiles. 111In-biotin nonspecifically accumulates at the inflammation site by increased capillary permeability. Approximately 4 hours after 111In-biotin is injected, avidin is administered via i.v. The 111In-biotin-avidin complex is formed, which quickly clears through the kidneys with minimal uptake by normal tissue. A high target-to-background ratio for early imaging is thus achieved. 111In-labeled biotin scintigraphy has been shown to be an important imaging method for spinal infections in patients.125–129 SPECT/CT increased the efficacy of 111In-biotin scintigraphy in the management of patients with spine infections.130
Vitamin B12
Vitamin B12 (cyanocobalamin) (Cbl) is a significant enzyme cofactor necessary for rapid replication of cells and bacteria.131 Vitamin B12 was labeled with either 99mTc or 111In via bifunctional chelating agents, and was evaluated to distinguish bacterial infection from sterile inflammation Initial results were highly encouraging.132
Chemotactic Peptides
Chemotactic compounds are attractants that are secreted by microorganisms to which are attracted white blood cells. This movement of an organism in response to a chemical stimulus is known as chemotaxis. In the case of pyogenic infection, PMNs are attracted to the various chemotactic stimuli released by the invading microorganism. One of the well-characterized stimuli is a peptide N-formyl-met-leu-phe (FMLP). With the hypothesis that radiolabeled FMLP, once administered intravenously, shall bind to circulating PMNs for convenient imaging of infection, Zoghbi et al133 labeled FMLP with 111In. Resultant neutropenia in a rabbit model prevented further investigations.
Ten years later, Fishman et al prepared FMLP analogs, labeled them with 99mTc. Further investigations in animals witnessed persistent neutropenia.134–137
Chemokines and Cytokines
Chemokines are signaling proteins secreted by cells. Their name is derived from their ability to induce directed chemotaxis in nearby responsive cells. Cytokine proteins are classified as kemokines according to their behavior and structural characteristics. Lymphocytic T cells and macrophages are main sources of cytokines. Cytokines are responsible for homeostatic control of the immune system. Cytokines interact with specific cell membrane receptors, which exist primarily on leukocytes. Their expression of cytokine receptors are increased in the course of infection and inflammation Radiolabeled cytokines with low molecular weight, short circulating time, high affinity for binding to the specific receptors, and human recombinant origin to prevent immunogenicity can in principle label WBCs in vivo.135,138–140
Tuftsin consists of an Fc portion of IgG. Tuftsin receptor is expressed on surface of neutrophils, monocytes, and macrophages, and it stimulates phagocytosis and chemotaxis by these cells.141 99mTc-RP128, a tuftsin receptor antagonist, demonstrated promising results for imaging inflammation in patients with Crohn’s disease and RA. No adverse events were reported.142–144
Interleukin-1 (IL-1) is a proinflammatory cytokine that binds to receptors expressed on granulocytes, monocytes, and lymphocytes. 123I-labeled IL-1 localized infectious foci in mice. In humans, however, serious adverse events prevented its further use.145–147
IL-2 is expressed on activated T lymphocytes. It binds to IL-2 receptors with high affinity, which are expressed by activated lymphocytes during inflammation. It is responsible for sustaining the inflammatory reaction with stimulation of long-term proliferation of activated T lymphocytes, and proliferation and differentiation of natural killer cells, B cell lymphocytes, and macrophages. 99mTc-or 123I-labeled IL-2 successfully showed T lymphocyte infiltration in various autoimmune and inflammatory diseases such as Hashimoto thyroiditis, Graves’ disease, Crohn’s disease, and celiac disease.148–153 99mTc-labeled IL-2 showed the presence of insulitis in patients with autoimmune diabetes.154 In the meantime, labeling IL-2 with 18F was advocated.3
IL-6 binds to specific receptors expressed on neutrophils. 125I-labeled IL-6 was used to image infection in mice. Accumulation of 125I-IL-6 was lower than 125I-IL-1.155
IL-8 is a chemotactic cytokine binds to CXC chemokine receptors on plasma membranes of neutrophils with high affinity It plays an important role in infection and inflammation It activates PMNs and provides their migration to inflammation or infection foci.156,157 125I-labeled recombinant human IL-8 (rhIL-8) localized acute inflammatory lesions in an animal model.158 131I-labeled rhIL-8 rapidly accumulated in lesions of osteomyelitis and cellulitis in patients.159 IL-8 labeled with 99mTc, using HYNIC as a chelator, was also evaluated to image infection in an animal model. It quickly visualized infection but produced transient neutropenia.160 99mTc-IL-8 scintigraphy was performed in patients with suspected, localized infections. No significant side effects were seen.161 However, no further data are found in the literature.
IL-12 stimulates the differentiation of T lymphocytes and plays a role in the chronic inflammation. IL-12 receptor is expressed on peripheral mononuclear cells and is present on activated T lymphocytes and natural killer cells.162,163 99mTc-HYNIC-IL-12 successfully imaged chemically induced chronic colitis in mice.164
Platelet factor 4 binds to CXC type II (IL-8 type B) receptors, which are expressed on PMNs and monocytes. P483H peptide, which includes the heparin-binding region of plate-let factor 4 was labeled with 99mTc and used to image Escherichia coli (E. coli) infections in rabbits and infection in patients. No adverse events were reported.165,166
Leukotriene B4 (LTB4) is a chemotactic factor and expressed at a high level by granulocytes.167 It was reported that radiolabeled LTB4 antagonist against LTB4 receptors can be used an agent to image inflammation and infection. The results of this agent were compared with various agents like 99mTc-HMPAO-labeled WBC. It was reported that DPC-11870 can clearly image abscesses. However, high-level distribution of this agent in bone marrow prevented its further use.168–170
Antimicrobial Peptides
Equally important to imaging bacterial infection is determining the effectiveness of antibiotic treatment so that any over treatment could be prevented. In this quest, targeting proliferating microorganism can play an important role. Attempts have been made to target bacteria using radiolabeled antimicrobial peptides, which are the protein molecules of the innate immune system to protect the body from infection.171 Antimicrobial peptides with their amphipathic structure pass through the membrane and enter inside the target bacterial cell.172 Interaction with the bacterial plasma membrane by electrostatic and hydrophobic interaction composes the main part of their antimicrobial activity. Therefore, radiolabeled antimicrobial peptides can in principle differentiate bacterial infection from sterile inflammation and can determine effectiveness of antibiotic treatment.173
Various radiolabeled antimicrobial peptides such as human neutrophil peptide, human lactoferrin, human ubiquicidin peptide fragment (UBI), and bacteriophages have been investigated for imaging of infection.174–178
99mTc-labeled human neutrophil peptide-1 rapidly imaged bacterial infections in mice.179 With a large protein such as 99mTc-labeled human lactoferrin, the imaging results, however, were not so preferable.173,180
Various domains of UBI such as UBI 29–41, UBI 18–35, and UBI 31–38 were labeled with 99mTc and investigated for imaging bacterial infections.173,176 99mTc-labeled UBI 29–41 differentiated bacterial and fungal infection from sterile inflammation in experimental animals and in humans.181–183 In a recent study, UBI fragments 29–41 and 31–38 were conjugated with NODAGA and labeled with 68Ga and investigated for imaging infection. Specifi uptake of the 68Ga-NODAGA-UBI conjugates was observed in S. aureus in mice by PET imaging.184
Bacteriophages185 P22, E79, VD-13, and 60 were labeled with 99mTc and evaluated in vitro and in an infection mouse model.186 Specific host binding was seen in vitro for all of the 99mTc-phages. Further investigations are warranted.
Annexin-V
Annexin is a common name for a group of cellular proteins. In humans, the annexins are found inside the cell. During bacterial invasion, PMNs are activated, migrate to the site of invasion, and phagocytose the microorganism. During the process, PMNs become apoptotic and externalize phosphatidylserine on their surface. Annexin-V, the endogenous protein, binds to phosphatidylserine with high affinity. With the hypothesis that radiolabeled Annexin-V selectively binds to apoptotic PMNs, Annexin-V was labeled with 99mTc and was successfully evaluated to image experimental abscesses in animals and infection in humans.187–190
Like Annexin-V near infrared fluorophore ophore, named PSVue794, also binds to phosphatidylserine with high affinity. Five microgram of PSVue794 was administered intravenously to mice bearing bacterial infection and sterile infland mice were imaged optically at various predetermined times after injection. At 3 hours post injection, abscesses were clearly visualized (abscess/background 6.6 ± 0.2) with greater uptake than in the inflammation (inflammation/ background 3.2 ± 0.5).191 The progressive and promising development in optical imaging technology, may in the future, make this non-nuclear technology feasible to image infection in humans.191,192
124I-FIAU Fialuridine
Fialuridine, 1-(2-deoxy-2-fluoro-β-D-arabinofuranosyl)-5-iodouracil (FIAU) is a thymidine nucleoside analog. Bacterial thymidine kinase (TK) uses FIAU as a substrate. The mechanism of uptake of FIAU is similar to uptake of FDG in that FIAU is phosphorylated and trapped inside of bacteria by TK.193–196 However, not all bacteria have TK enzyme, which might limit its applications.196
Nonetheless, Bettegowda et al developed a technique to label FIAU with 125I and demonstrated focal infection in a mouse model. The investigators concluded that this agent can be used for focal bacterial infections in humans.194
Diaz et al labeled FIAU with positron emitting 124I for PET musculoskeletal bacterial infection in a group of 8 patients.195 Encouraging results of this work prompted Zhang et al to image prosthetic joint infection in 6 patients. Results were similar to those of 111In-WBC. The image quality, however, was inferior to that with 111In-WBC. Although qualifies for PET imaging, the 5.4 d t1/2 of 124I may be considered too long for a diagnostic application of the radionuclide.196
99mTc-Pheophorbide-Α
Pheophorbide-α (PH-A) is a plant derivative photosensitizing agent, extracted from spirulina maxima algae. PH-A has been shown to have microbial activity both for gram (+) and gram (−) bacteria.
99mTc-labeled PH-A was investigated to image S. aureus infection in rats with 99mTc-PH-A. Target-to-nontarget radioactivity ratio was significantly higher than that of 99mTc-labeled antibiotics. The authors concluded that 99mTc-PH-A can be developed as a radiopharmaceutical to discriminate bacterial infection from inflammation.197
18F-Labeled Maltohexaose
18F-labeled maltohexaose (18F-MH) is a novel PET imaging agent that targets bacteria-specific maltodextrin transporter not found in mammalian cells. This makes 18F-MH specific for viable bacteria and allows quick clearance from noninfected tissues. This, in turn, renders 18F-MH an agent with higher sensitivity and specificity than 18FDG to image pyogenic infection. As a commonly used food additive, 18F-MH has minimal toxicity in humans.
Consistent with this hypothesis, Ning et al showed 18F-MH can image early-stage infections consisting of as few as 105 colony forming unit E. coli in rats. It may be possible that 18F-MH can determine drug resistance and allow physicians to monitor the course of antibiotic treatment. Further work is warranted.198
(1→ 3)-β-D-Glucan Aptamers
Glucans are found in cell membrane of some bacteria. Aptamers similar to antibodies have high affinity and specificity for glucans. Unlike antibodies, however, they are minimally immunogenic, and relatively inexpensive to synthesize and modify. Their small size allows them to penetrate into cells. Many radiolabeled aptamers have been used to image inflammation and infection in preclinical studies.199–201
Santos et al evaluated 99mTc-labeled anti-S. aureus aptamers to detect bacterial infection. The 99mTc-radiolabeled aptamers were able to differentiate aseptic inflammation from bacterial infection.202
In another study, 99mTc-labeled (1→ 3)-β-D-glucan aptamers were evaluated to identity foci of fungal infection. The authors concluded that the use of aptamers for scintigraphic imaging of bacterial infections should be further investigated.203
Siderophores
Siderophores are iron transporters used by most living cells, including bacteria, and serve as a chelating agent for such metal ions as gallium or indium.
Siderophores such as triacetylfusarinine, ferrioxamine E, desferrichrome A, and desferrioxamine have been labeled with 68Ga and 89Zr, and compared for imaging Aspergillus infection.204 All of radiolabeled siderophores showed promising properties as radiopharmaceuticals for PET imaging of infection.205
Summary and Future Directions
Infection is ubiquitous yet heterogeneous. Formation of abscesses is a highly coordinated and a complex set of biological, biochemical, and pathologic events that protect a body from pathogenic invasion. One avenue that helps the physician in the management of this disease is the scintigraphic localization of occult abscesses. Investigations into approaches for imaging infection begun nearly half a century ago. However, the quest for finding an ideal agent to image infection continues until today.
The investigations to image infection began with a search for a radioactive agent that will label WBC effectively, yet preserve their viability, so that the labeled WBCs shall transport the radioactivity to the site of infection and allow it to be localized by external scintigraphic imaging. Although this approach has been highly successful in principle, it is far from ideal, primarily because of the processes that are required to label WBCs ex vivo. These weaknesses combined with the increased understanding of pathologic response to invading microorganism, at a cellular and a molecular level, have paved the way to the scholarly approaches for the preparation and evaluation of numerous radioactive agents to image infection. Many of these are listed in this review with their pros and cons together with a few, such as 99mTc-HMPAO-labeled autologous WBC, 111In-oxine-labeled autologous WBC, 18F-FDG, and 67Ga-citrate that are used in routine practice worldwide. Do they address all questions that need to be addressed for the management of patients with infection? No. Are they ideal for the routine clinical use? No. The questions then cross the mind of all those basic scientists and physicians who are closely involved with the fi are (1) Will a single ideal agent ever be discovered that shall image all heterogeneous types of infections conveniently and reliably? (2) Will one require to use multiple agents that may be most suitable to localize a given type of infection and be able to distinguish it from inflammation or (3) Will the imaging examination performed using 1 agent shall permit physicians to prevent overtreatment of a patient? Looking closely at the half century old history of the development of the field, it is reasonable to conclude that the choice of the use of a particular radiopharmaceutical shall be governed by the convenience of its use and by the ability of a given radiopharmaceutical to address a specific question that may be needed to address for better management of the disease. We hope that this presentation providing a description of multiple radiopharmaceuticals with the underlying principals of their use and pros and cons of their applications shall be helpful in the future, in making such a choice.
Acknowledgment
The principal author (MLT) thanks Dr. Christopher Palestro, the Editor of this special issue, for giving him the opportunity to write this review article and his colleagues for their contribution. Dr. Ebru Salmanoglu is a visiting scholar from The Scientific and Technological Research Council of Turkey (TÜBİTAK), to the Thakur laboratories, and gratefully acknowledges the fellowship from the Turkish The Scientific and Technological Research Council of Turkey (TÜBỈTAK).
References
- 1.Kakkar D, Tiwari AK, Singh H, et al. : Past and present scenario of imaging infection and inflammation: A nuclear medicine perspective. Mol Imaging 11:309–337, 2012 [Google Scholar]
- 2.Das SS, Hall AV, Wareham DW, et al. : Infection imaging with radiopharmaceuticals in the 21st century. Braz arch biol technol 45:25–37, 2002 [Google Scholar]
- 3.Signore A, Glaudemans AW: The molecular imaging approach to image infections and inflammation by nuclear medicine techniques. Ann Nucl Med 25:681–700, 2011 [DOI] [PubMed] [Google Scholar]
- 4.Goldsmith SJ, Vallabhajosula S: Clinically proven radiopharmaceuticals for infection imaging: Mechanisms and applications. Semin Nucl Med 39:2–10, 2009 [DOI] [PubMed] [Google Scholar]
- 5.Petruzzi N, Shanthly N, Thakur M: Recent trends in soft-tissue infection imaging. Semin Nucl Med 39:115–123, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Boerman OC, Rennen H, Oyen WJ, et al. : Radiopharmaceuticals to image infection and inflammation. Semin Nucl Med 31:286–295, 2001 [DOI] [PubMed] [Google Scholar]
- 7.Lavender JP, Lowe J, Barker JR, et al. : Gallium 67 citrate scanning in neoplastic and inflammatory lesions. Br J Radiol 44:361–366, 1971 [DOI] [PubMed] [Google Scholar]
- 8.McAfee JG, Thakur ML: Survey of radioactive agents for in vitro labeling of phagocytic leukocytes. I. Soluble agents. J Nucl Med 17:480–487, 1976 [PubMed] [Google Scholar]
- 9.McAfee JG, Thakur ML: Survey of radioactive agents for in vitro labeling of phagocytic leukocytes. II. Particles. J Nucl Med 17:488–492, 1976 [PubMed] [Google Scholar]
- 10.Thakur ML, Coleman RE, Mayhall CG, et al. : Preparation and evaluation of 111In-labeled leukocytes as an abscess imaging agent in dogs. Radiology 119:731, 1976 [DOI] [PubMed] [Google Scholar]
- 11.Thakur ML, Lavender JP, Arnot RN, et al. : Indium-111-labeled autologous leukocytes in man. J Nucl Med 18:1014–1021, 1977 [PubMed] [Google Scholar]
- 12.Connolly CM, Donohoe KJ: Nuclear medicine imaging of infection. Semin Roentgenol 52:114–119, 2017 [DOI] [PubMed] [Google Scholar]
- 13.Thakur ML, McKenney SL, Park CH: Evaluation of indium-111–2-mercaptopyridine-N-oxide for labeling leukocytes in plasma: A kit preparation. J Nucl Med 26:518–523, 1985 [PubMed] [Google Scholar]
- 14.Intenzo CM, Desai AG, Thakur ML, et al. : Comparison of leukocytes labeled with indium-111–2-mercaptopyridine-N-oxide and indium-111 oxine for abscess detection. J Nucl Med 28:438–441, 1987 [PubMed] [Google Scholar]
- 15.Schauwecker DS, Burt RW, Park HM, et al. : Clinical comparison of indium-111 acetylacetone and indium-111 tropolone granulocytes. J Nucl Med 27:1675–1679, 1986 [PubMed] [Google Scholar]
- 16.Peters AM, Roddie ME, Danpure HJ, et al. : 99Tcm-HMPAO labelled leucocytes: Comparison with 111In-tropolonate labelled granulocytes. Nucl Med Commun 9:449–463, 1988 [DOI] [PubMed] [Google Scholar]
- 17.Peters AM, Danpure HJ, Osman S, et al. : Clinical experience with 99mTc-hexamethylpropylene-amineoxime for labelling leucocytes and imaging inflammation. Lancet 2:946–949, 1986 [DOI] [PubMed] [Google Scholar]
- 18.Robins PD, Salazar I, Forstrom LA, et al. : Biodistribution and radiation dosimetry of stabilized 99mTc-exametazine-labeled leukocytes in normal subjects. J Nucl Med 41:934–940, 2000 [PubMed] [Google Scholar]
- 19.Weldon MJ, Joseph AE, French A, et al. : Comparison of 99m technetium hexamethylpropylene-amine oxime labelled leucocyte with 111-indium tropolonate labelled granulocyte scanning and ultrasound in the diagnosis of intra-abdominal abscess. Gut 37:557–564, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Erba PA, Glaudemans AW, Veltman NC, et al. : Image acquisition and interpretation criteria for 99mTc-HMPAO-labelled white blood cell scintigraphy: Results of a multicentre study. Eur J Nucl Med Mol Imaging 41:615–623, 2014 [DOI] [PubMed] [Google Scholar]
- 21.He YX, Guo QY: Clinical applications and advances of positron emission tomography with fluorine-18-fluorodeoxyglucose (18F-FDG) in the diagnosis of liver neoplasms. Postgrad Med J 84:246–251, 2008 [DOI] [PubMed] [Google Scholar]
- 22.Wu C, Li F, Niu G, et al. : PET imaging of inflammation biomarkers. Theranostics 3:448–466, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Autio A, Jalkanen S, Roivainen A: Nuclear imaging of inflammation: Homing-associated molecules as targets. EJNMMI Res 3:1, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rini JN, Bhargava KK, Tronco GG, et al. : PET with FDG-labeled leukocytes versus scintigraphy with 111In-oxine-labeled leukocytes for detection of infection. Radiology 238:978–987, 2006 [DOI] [PubMed] [Google Scholar]
- 25.Palestro CJ, Love C, Bhargava KK: Labeled leukocyte imaging: Current status and future directions. Q J Nucl Med Mol Imaging 53:105–123, 2009 [PubMed] [Google Scholar]
- 26.Bhargava KK, Gupta RK, Love C, et al. : Human leukocyte labeling with 64Cu: An intraindividual comparison with 111In-oxine and 18F-FDG. J Nucl Med 49:133P, 2008. (suppl 1) [Google Scholar]
- 27.Jamar F, Buscombe J, Chiti A, et al. : EANM/SNMMI guideline for 18F-FDG use in inflammation and infection. J Nucl Med 54:647–658, 2013 [DOI] [PubMed] [Google Scholar]
- 28.Ahmed FZ, James J, Memmott MJ, et al. : Radionuclide imaging of cardiovascular infection. Cardiol Clin 34:149–165, 2016 [DOI] [PubMed] [Google Scholar]
- 29.Glaudemans AW, de Vries EF, Galli F, et al. : The use of (18)F-FDG-PET/CT for diagnosis and treatment monitoring of inflammatory and infectious diseases. Clin Dev Immunol 2013:623036, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gotthardt M, Bleeker-Rovers CP, Boerman OC, et al. : Imaging of inflammation by PET, conventional scintigraphy, and other imaging techniques. J Nucl Med Technol 41:157–169, 2013 [DOI] [PubMed] [Google Scholar]
- 31.Manohar K, Mittal BR, Jain S, et al. : F-18 FDG-PET/CT in evaluation of patients with fever of unknown origin. Jpn J Radiol 31:320–327, 2013 [DOI] [PubMed] [Google Scholar]
- 32.Balink H, Collins J, Bruyn GA, et al. : F-18 FDG PET/CT in the diagnosis of fever of unknown origin. Clin Nucl Med 34:862–868, 2009 [DOI] [PubMed] [Google Scholar]
- 33.Jasper N, Däbritz J, Frosch M, et al. : Diagnostic value of [(18)F]-FDG PET/CT in children with fever of unknown origin or unexplained signs of inflammation Eur J Nucl Med Mol Imaging 37:136–145, 2010 [DOI] [PubMed] [Google Scholar]
- 34.Bhargava KK, Gupta RK, Nichols KJ, et al. : In vitro human leukocyte labeling with (64)Cu: An intraindividual comparison with (111)In-oxine and (18)F-FDG. Nucl Med Biol 36:545–549, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fairclough M, Prenant C, Ellis B, et al. : A new technique for the radiolabelling of mixed leukocytes with zirconium-89 for inflammation imaging with positron emission tomography. J Labelled Comp Radiopharm 59:270–276, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sato N, Wu H, Asiedu KO, et al. : (89)Zr-Oxine complex PET cell imaging in monitoring cell-based therapies. Radiology 275:490–500, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Park JA, Lee YJ, Lee JW, et al. : Evaluation of [(89)Zr]-oxalate as a PET tracer in inflammation, tumor, and rheumatoid arthritis models. Mol Pharm 13:2571–2577, 2016 [DOI] [PubMed] [Google Scholar]
- 38.Locher JT, Seybold K, Andres RY, et al. : Imaging of inflammatory and infectious lesions after injection of radioiodinated monoclonal anti-granulocytes antibodies. Nucl Med Commun 7:659–670, 1986 [PubMed] [Google Scholar]
- 39.Buchegger F, Schreyer M, Carrel S, et al. : Monoclonal antibodies identify a CEA crossreacting antigen of 95 kD (NCA-95) distinct in antigenicity and tissue distribution from the previously described NCA of 55 kD. Int J Cancer 33:643–649, 1984 [DOI] [PubMed] [Google Scholar]
- 40.Becker W, Bair J, Behr T, et al. : Detection of soft-tissue infections and osteomyelitis using a technetium-99m-labeled anti-granulocyte monoclonal antibody fragment. J Nucl Med 35:1436–1443, 1994 [PubMed] [Google Scholar]
- 41.Becker W, Laurer U, Marienhagen J, et al. : Comparison of Tc-99m-antigranulocyte (NCA-95) Fab’ fragments and scintigraphy with In-111-oxine/Tc99m-HMPAO-labelled leukocytes in infectious diseases. J Nucl Med 33:903, 1992 [Google Scholar]
- 42.Corstens FH, Oyen WJ, Becker WS: Radioimmunoconjugates in the detection of infection and inflammation Semin Nucl Med 23:148–164, 1993 [DOI] [PubMed] [Google Scholar]
- 43.Bosslet K, Lüben G, Schwarz A, et al. : Immunohistochemical localization and molecular characteristics of three monoclonal antibody-defined epitopes detectable on carcinoembryonic antigen (CEA). Int J Cancer 36:75–84, 1985 [DOI] [PubMed] [Google Scholar]
- 44.Bosslet K, Steinsträser A, Schwarz A, et al. : Generation and functional characteristics of the granulocyte selective monoclonal antibody BW 250/183. Nucl Med 28:151, 1989 [Google Scholar]
- 45.Thakur ML, Richard MD, White FW 3rd: Monoclonal antibodies as agents for selective radiolabeling of human neutrophils. J Nucl Med 29:1817–1825, 1988 [PubMed] [Google Scholar]
- 46.Thakur ML, Marcus CS, Henneman P, et al. : Imaging inflammatory diseases with neutrophil-specific technetium-99m-labeled monoclonal antibody anti-SSEA-1. J Nucl Med 37:1789–1795, 1996 [PubMed] [Google Scholar]
- 47.Mozley PD, Thakur ML, Alavi A, et al. : Effects of a 99mTc-labeled murine immunoglobulin M antibody to CD15 antigens on human granulocyte membranes in healthy volunteers. J Nucl Med 40:2107–2114, 1999 [PubMed] [Google Scholar]
- 48.John E, Thakur ML, Wilder S, et al. : Technetium-99m-labeled monoclonal antibodies: Influence of technetium-99m binding sites. J Nucl Med 35:876–881, 1994 [PubMed] [Google Scholar]
- 49.Love C, Tronco GG, Palestro CJ: Imaging of infection and inflammation with 99mTc-Fanolesomab. Q J Nucl Med Mol Imaging 50:113–120, 2006 [PubMed] [Google Scholar]
- 50.Tronco GG, Love C, Rini JN, et al. : Diagnosing prosthetic vascular graft infection with the antigranulocyte antibody 99mTc-fanolesomab. Nucl Med Commun 28:297–300, 2007 [DOI] [PubMed] [Google Scholar]
- 51.Shanthly N, Aruva MR, Zhang K, et al. : 99mTc-Fanolesomab: Affinity, pharmacokinetics and preliminary evaluation. Q J Nucl Med Mol Imaging 50:104–112, 2006 [PubMed] [Google Scholar]
- 52.Lucignani G: The many roads to infection imaging. Eur J Nucl Med Mol Imaging 34:1873–1877, 2007 [DOI] [PubMed] [Google Scholar]
- 53.Kumar V: Radiolabeled white blood cells and direct targeting of micro-organisms for infection imaging. Q J Nucl Med Mol Imaging 49:325–338, 2005 [PubMed] [Google Scholar]
- 54.Glaudemans AW, Dierckx RA, Kallenberg CG, et al. : The role of radiolabelled anti-TNFa monoclonal antibodies for diagnostic purposes and therapy evaluation. Q J Nucl Med Mol Imaging 54:639–653, 2010 [PubMed] [Google Scholar]
- 55.Scott DL, Kingsley GH: Tumour necrosis factor inhibitors for rheumatoid arthritis. N Engl J Med 355:704–712, 2006 [DOI] [PubMed] [Google Scholar]
- 56.Deleuran BW, Chu CQ, Field M, et al. : Localization of tumor necrosis factor receptors in the synovial tissue and cartilage-pannus junction in patients with rheumatoid arthritis. Implications for local actions of tumor necrosis factor alpha. Arthritis Rheum 35:1170–1178, 1992 [DOI] [PubMed] [Google Scholar]
- 57.Olsen NJ, Stein CM: New drugs for rheumatoid arthritis. N Engl J Med 350:2167–2179, 2004 [DOI] [PubMed] [Google Scholar]
- 58.Alstergren P, Larsson PT, Kopp S: Successful treatment with multiple intra-articular injections of infliximab in a patient with psoriatic arthritis. Scand J Rheumatol 37:155–157, 2008 [DOI] [PubMed] [Google Scholar]
- 59.Boesen M, Boesen L, Jensen KE, et al. : Clinical outcome and imaging changes after intraarticular (IA) application of etanercept or methylprednisolone in rheumatoid arthritis: Magnetic resonance imaging and ultrasound-Doppler show no effect of IA injections in the wrist after 4 weeks. J Rheumatol 35:584–591, 2008 [PubMed] [Google Scholar]
- 60.Haroon M, O’Gradaigh D: Effi and safety of combining intra-articular methylprednisolone and anti-TNF agent to achieve prolonged remission in patients with recurrent inflammatory monoarthritis. Joint Bone Spine 77:232–234, 2010 [DOI] [PubMed] [Google Scholar]
- 61.Ceccarelli F, Perricone C, Galli F, et al. : Use of 99mTc-labelled anti-TNF monoclonal antibodies to assess patients affected by inflammatory arthropathies. Int J Radiol Med Imag 1:102, 2015 [Google Scholar]
- 62.Conti F, Priori R, Chimenti MS, et al. : Successful treatment with intraarticular infliximab for resistant knee monoarthritis in a patient with spondyloarthropathy: A role for scintigraphy with 99mTc-infliximab. Arthritis Rheum 52:1224–1246, 2005 [DOI] [PubMed] [Google Scholar]
- 63.Conti F, Malviya G, Ceccarelli F, et al. : Role of scintigraphy with 99mTc-infliximab in predicting the response of intraarticular infliximab treatment in patients with refractory monoarthritis. Eur J Nucl Med Mol Imaging 39:1339–1347, 2012 [DOI] [PubMed] [Google Scholar]
- 64.Malviya G, Conti F, Chianelli M, et al. : Molecular imaging of rheumatoid arthritis by radiolabelled monoclonal antibodies: New imaging strategies to guide molecular therapies. Eur J Nucl Med Mol Imaging 37:386–398, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Barrera P, Oyen WJ, Boerman OC, et al. : Scintigraphic detection of tumour necrosis factor in patients with rheumatoid arthritis. Ann Rheum Dis 62:825–828, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Malviya G, D’Alessandria C, Lanzolla T, et al. : 99mTechnetium labelled anti-TNF-α antibodies for the therapy decision-making and follow-up of patients with rheumatoid arthritis. QJ Nucl Med Mol Imaging 52:13–14, 2008. (suppl 1) [Google Scholar]
- 67.Cinader B, Miller RG (eds): Progress in Immunology VI: Sixth International Congress of Immunology Orlando, Florida, Academic Press, 1986 [Google Scholar]
- 68.Becker W, Emmrich F, Horneff G, et al. : Imaging rheumatoid arthritis specifically with technetium 99m CD4-specific (T-helper lymphocytes) antibodies. Eur J Nucl Med 17:156–159, 1990 [DOI] [PubMed] [Google Scholar]
- 69.Steinhoff K, Pierer M, Siegert J, et al. : Visualizing inflammation activity in rheumatoid arthritis with Tc-99 m anti-CD4-mAb fragment scintigraphy. Nucl Med Biol 41:350–354, 2014 [DOI] [PubMed] [Google Scholar]
- 70.Stalteri MA, Mather SJ: Technetium-99m labelling of the anti-tumour antibody PR1A3 by photoactivation. Eur J Nucl Med 23:178–187, 1996 [DOI] [PubMed] [Google Scholar]
- 71.Malviya G, Laganà B, Milanetti F, et al. : Use of 99m-technetium labelled Rituximab for imaging of patients with chronic inflammatory diseases. Eur J Nucl Med Mol Imaging 35:S142–S143, 2008 [Google Scholar]
- 72.Marcus C, Thakur ML, Huynh TV, et al. : Imaging rheumatic joint diseases with anti-T lymphocyte antibody OKT-3. Nucl Med Commun 15:824–830, 1994 [DOI] [PubMed] [Google Scholar]
- 73.Martins FP, Gutfilen B, de Souza SA, et al. : Monitoring rheumatoid arthritis synovitis with 99mTc-anti-CD3. Br J Radiol 81:25–29, 2008 [DOI] [PubMed] [Google Scholar]
- 74.Malviya G, De Vries EFJ, Dierckx RA, et al. : Radiopharmaceuticals for imaging chronic lymphocytic inflammation. Braz arch biol technol 50:1–13, 2007 [Google Scholar]
- 75.Malviya G, D’Alessandria C, Trotta C, et al. : Radiolabeled-Visilizumab, a humanized anti-CD3 monoclonal antibody, for in vivo targeting of human CD3+ lymphocytes. Eur J Nucl Med Mol Imaging 35:S142, 2008. (suppl 2) [DOI] [PubMed] [Google Scholar]
- 76.Bhatti M, Chapman P, Peters M, et al. : Visualising E-selectin in the detection and evaluation of infl y bowel disease. Gut 43:40–47, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jamar F, Chapman PT, Manicourt DH, et al. : A comparison between 111In-anti-E-selectin mAb and 99Tcm-labelled human non-specific immunoglobulin in radionuclide imaging of rheumatoid arthritis. Br J Radiol 70:473–481, 1997 [DOI] [PubMed] [Google Scholar]
- 78.Jamar F, Houssiau FA, Devogelaer JP, et al. : Scintigraphy using a technetium 99m-labelled anti-E-selectin Fab fragment in rheumatoid arthritis. Rheumatology (Oxford) 41:53–61, 2002 [DOI] [PubMed] [Google Scholar]
- 79.Keelan ET, Harrison AA, Chapman PT, et al. : Imaging vascular endothelial activation: An approach using radiolabeled monoclonal antibodies against the endothelial cell adhesion molecule E-selectin. J Nucl Med 35:276–281, 1994 [PubMed] [Google Scholar]
- 80.Buscombe J: Radiolabelled human immunoglobulins. Nucl Med Commun 16:990–1001, 1995 [DOI] [PubMed] [Google Scholar]
- 81.de Kleijn EM, Oyen WJ, Corstens FH, et al. : Utility of indium-111-labeled polyclonal immunoglobulin G scintigraphy in fever of unknown origin. The Netherlands FUO Imaging Group. J Nucl Med 38:484–489, 1997 [PubMed] [Google Scholar]
- 82.Gerasimou G, Moralidis E, Papanastasiou E, et al. : Radionuclide imaging with human polyclonal immunoglobulin (Tc-HIG) and bone scan in patients with rheumatoid arthritis and serum-negative polyarthritis. Hippokratia 15:37–42, 2011 [PMC free article] [PubMed] [Google Scholar]
- 83.Lambrecht FY: Evaluation of 99(m)Tc-labeled antibiotics for infection detection. Ann Nucl Med 25:1–6, 2011 [DOI] [PubMed] [Google Scholar]
- 84.Auletta S, Galli F, Lauri C, et al. : Imaging bacteria with radiolabelled quinolones, cephalosporins and siderophores for imaging infection: A systematic review. Clin Transl Imaging 4:229–252, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Vinjamuri S, Hall AV, Solanki KK, et al. : Comparison of 99mTc infecton imaging with radiolabelled white-cell imaging in the evaluation of bacterial infection. Lancet 347:233–235, 1996 [DOI] [PubMed] [Google Scholar]
- 86.von Rosenstiel N, Adam D: Quinolone antibacterials. An update of their pharmacology and therapeutic use. Drugs 47:872–901, 1994 [DOI] [PubMed] [Google Scholar]
- 87.Jacoby GA: Mechanisms of resistance to quinolones. Clin Infect Dis 41:S120–S126, 2005. (suppl 2) [DOI] [PubMed] [Google Scholar]
- 88.Britton KE, Vinjamuri S, Hall AV, et al. : Clinical evaluation of technetium-99m infecton for the localisation of bacterial infection. Eur J Nucl Med 24:553–556, 1997 [DOI] [PubMed] [Google Scholar]
- 89.Satpati D, Arjun C, Krishnamohan R, et al. : (68) Ga-labeled ciprofloxacin conjugates as radiotracers for targeting bacterial infection. Chem Biol Drug Des 87:680–686, 2016 [DOI] [PubMed] [Google Scholar]
- 90.Brunner M, Langer O, Dobrozemsky G, et al. : [18F]Ciprofloxacin, a new positron emission tomography tracer for noninvasive assessment of the tissue distribution and pharmacokinetics of ciprofloxacin in humans. Antimicrob Agents Chemother 48:3850–3857, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.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 32:143–150, 2005 [DOI] [PubMed] [Google Scholar]
- 92.Britton KE, Wareham DW, Das SS, et al. : Imaging bacterial infection with (99m)Tc-ciprofl (Infecton). J Clin Pathol 55:817–823, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Lee M, Yoon M, Hwang KH, et al. : Tc-99m Ciprofloxacin SPECT of pulmonary tuberculosis. Nucl Med Mol Imaging 44:116–122, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Malamitsi J, Papadopoulos A, Vezyrgianni A, et al. : 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 32:1060–1069, 2011 [DOI] [PubMed] [Google Scholar]
- 95.Fuster D, Soriano A, Garcia S, et al. : Usefulness of 99mTc-ciprofloxacin scintigraphy in the diagnosis of prosthetic joint infections. Nucl Med Commun 32:44–51, 2011 [DOI] [PubMed] [Google Scholar]
- 96.Falagas ME, Valotassiou VJ, Papadouli D, et al. : 99mTechnetiumciprofloxacin scintigraphy for the evaluation of spinal infections: A preliminary report. Clin Orthop Relat Res 444:34–37, 2006 [DOI] [PubMed] [Google Scholar]
- 97.Bhardwaj V, Agrawal M, Suri T, et al. : Evaluation of adequacy of short-course chemotherapy for extraspinal osteoarticular tuberculosis using 99mTc ciprofloxacin scan. Int Orthop 35:1869–1874, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Shah SQ, Khan AU, Khan MR: Radiosynthesis and biodistribution of (99m)Tc-rifampicin: A novel radiotracer for in-vivo infection imaging. Appl Radiat Isot 68:2255–2260, 2010 [DOI] [PubMed] [Google Scholar]
- 99.Shah SQ, Khan AU, Khan MR: Radiosynthesis of 99mTc-nitrofurantoin a novel radiotracer for in vivo imaging of Escherichia coli infection. J Radioanal Nucl Chem 287:417–422, 2011 [Google Scholar]
- 100.Shahzadi SK, Qadir MA, Shahzad S, et al. : 99mTc-amoxicillin: A novel radiopharmaceutical for infection imaging. Arabian J Chem 2015, doi: 10.1016/j.arabjc.2015.04.003 [DOI] [Google Scholar]
- 101.Tsopelas C, Penglis S, Ruszkiewicz A, et al. : 99mTc-alafosfalin: An antibiotic peptide infection imaging agent. Nucl Med Biol 30:169–175, 2003 [DOI] [PubMed] [Google Scholar]
- 102.Martínez ME, Kiyono Y, Noriki S, et al. : New radiosynthesis of 2-deoxy-2-[(18)F]fluoroacetamido-D-glucopyranose and its evaluation as a bacterial infections imaging agent. Nucl Med Biol 38:807–817, 2011 [DOI] [PubMed] [Google Scholar]
- 103.Roohi S, Mushtaq A, Malik SA: Synthesis and biodistribution of 99mTc-Vancomycin in a model of bacterial infection. Radiochim Acta 93:415–418, 2005 [Google Scholar]
- 104.Roohi S, Mushtaq A, Jehangir M, et al. : Synthesis, quality control and biodistribution of 99mTc-Kanamycin. J Radioanal Nucl Chem 267:561–566, 2006 [Google Scholar]
- 105.Abdel-Ghaney IY, Sanad MH: Synthesis of 99mTc-erythromycin complex as a model for infection sites imaging. Radiochem 55:418–422, 2013 [Google Scholar]
- 106.Sanad MH: Labeling and biological evaluation of 99mTc-azithromycin for infective inflammation diagnosis. Radiochem 55:539–544, 2013 [Google Scholar]
- 107.Borai EH, Sanad MH, Fouzy ASM: Optimized chromatographic separation and biological evaluation of 99mTc-Clarithromycin for infective inflammation diagnosis. Radiochem 58:84–91, 2016 [Google Scholar]
- 108.Hina S, Rajoka MI, Roohi S, et al. : Preparation, biodistribution, and scintigraphic evaluation of (99m)Tc-clindamycin: An infection imaging agent. Appl Biochem Biotechnol 174:1420–1433, 2014 [DOI] [PubMed] [Google Scholar]
- 109.Inceboz T, Lambrecht FY, Yilmaz O, et al. : Technetium-99m labeled Mebendazole and biodistribution in experimentally Trichinella spiralis-infected rats. Med Chem Re 21:804–809, 2012 [Google Scholar]
- 110.Motaleb MA, Alabdullah ES, Zaghary WA: Synthesis, radiochemical and biological characteristics of 99mTc-8-hydroxy-7-substituted quinoline complex: A novel agent for infection imaging. J Radioanal Nucl Chem 287:61–67, 2011 [Google Scholar]
- 111.Lupetti A, de Boer MG, Erba P, et al. : Radiotracers for fungal infection imaging. Med Mycol 49:S62–S69, 2011. (suppl 1) [DOI] [PubMed] [Google Scholar]
- 112.Lupetti A, Welling MM, Mazzi U, et al. : Technetium-99m labelled fluconazole and antimicrobial peptides for imaging of Candida albicans and Aspergillus fumigatus infections. Eur J Nucl Med Mol Imaging 29:674–679, 2002 [DOI] [PubMed] [Google Scholar]
- 113.Siaens R, Eijsink VG, Vaaje-Kolstad G, et al. : Synthesis and evaluation of a 99mTechnetium labeled chitin-binding protein as potential specifi radioligand for the detection of fungal infections in mice. Q J Nucl Med Mol Imaging 50:155–166, 2006 [PubMed] [Google Scholar]
- 114.Siaens R, Eijsink VG, Dierckx R, et al. : (123)I-Labeled chitinase as specific radioligand for in vivo detection of fungal infections in mice. J Nucl Med 45:1209–1216, 2004 [PubMed] [Google Scholar]
- 115.Gabizon A, Catane R, Uziely B, et al. : Prolonged circulation time and enhanced accumulation in malignant exudates of doxorubicin encapsulated in polyethylene-glycol coated liposomes. Cancer Res 54:987–992, 1994 [PubMed] [Google Scholar]
- 116.Oyen WJ, Boerman OC, Storm G, et al. : Detecting infection and inflammation with technetium-99m-labeled Stealth liposomes. J Nucl Med 37:1392–1397, 1996 [PubMed] [Google Scholar]
- 117.Oyen WJ, Boerman OC, Dams ET, et al. : Scintigraphic evaluation of experimental colitis in rabbits. J Nucl Med 38:1596–1600, 1997 [PubMed] [Google Scholar]
- 118.Szebeni J: The interaction of liposomes with the complement system. Crit Rev Ther Drug Carrier Syst 15:57–88, 1998 [PubMed] [Google Scholar]
- 119.Koukourakis MI, Koukouraki S, Giatromanolaki A, et al. : Liposomal doxorubicin and conventionally fractionated radiotherapy in the treatment of locally advanced non-small-cell lung cancer and head and neck cancer. J Clin Oncol 17:3512–3521, 1999 [DOI] [PubMed] [Google Scholar]
- 120.Dams ET, Oyen WJ, Boerman OC, et al. : 99mTc-PEG liposomes for the scintigraphic detection of infection and inflammation: Clinical evaluation. J Nucl Med 41:622–630, 2000 [PubMed] [Google Scholar]
- 121.Brouwers AH, De Jong DJ, Dams ET, et al. : Tc-99m-PEG-Liposomes for the evaluation of colitis in Crohn’s disease. J Drug Target 8:225–233, 2000 [DOI] [PubMed] [Google Scholar]
- 122.Streule K, de Schrijver M, Fridrich R: 99Tcm-labelled HSA-nanocolloid versus 111In oxine-labelled granulocytes in detecting skeletal septic process. Nucl Med Commun 9:59–67, 1988 [PubMed] [Google Scholar]
- 123.De Schrijver M, Streule K, Senekowitsch R, et al. : Scintigraphy of inflammation with nanometer-sized colloidal tracers. Nucl Med Commun 8:895–908, 1987 [DOI] [PubMed] [Google Scholar]
- 124.Goins BA: Radiolabeled lipid nanoparticles for diagnostic imaging. Expert Opin Med Diagn 2:853–873, 2008 [DOI] [PubMed] [Google Scholar]
- 125.Rusckowski M, Paganelli G, Hnatowich DJ, et al. : Imaging osteomyelitis with streptavidin and indium-111-labeled biotin. J Nucl Med 37:1655–1662, 1996 [PubMed] [Google Scholar]
- 126.Lazzeri E, Manca M, Molea N, et al. : Clinical validation of the avidin/ indium-111 biotin approach for imaging infection/inflammation in orthopaedic patients. Eur J Nucl Med 26:606–614, 1999 [DOI] [PubMed] [Google Scholar]
- 127.Lazzeri E, Pauwels EK, Erba PA, et al. : Clinical feasibility of two-step streptavidin/111In-biotin scintigraphy in patients with suspected vertebral osteomyelitis. Eur J Nucl Med Mol Imaging 31:1505–1511, 2004 [DOI] [PubMed] [Google Scholar]
- 128.Attwood PV: The structure and the mechanism of action of pyruvate carboxylase. Int J Biochem Cell Biol 27:231–249, 1995 [DOI] [PubMed] [Google Scholar]
- 129.Lazzeri E, Erba P, Perri M, et al. : Scintigraphic imaging of vertebral osteomyelitis with 111in-biotin. Spine 33:E198–E204, 2008 [DOI] [PubMed] [Google Scholar]
- 130.Lazzeri E, Erba P, Perri M, et al. : Clinical impact of SPECT/CT with In-111 biotin on the management of patients with suspected spine infection. Clin Nucl Med 35:12–17, 2010 [DOI] [PubMed] [Google Scholar]
- 131.Rodionov DA, Vitreschak AG, Mironov AA, et al. : Comparative genomics of the vitamin B12 metabolism and regulation in prokaryotes. J Biol Chem 278:41148–41159, 2003 [DOI] [PubMed] [Google Scholar]
- 132.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. Mol Imaging Biol 17:829–837, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zoghbi SS, Thakur ML, Gottschalk A, et al. : Selective cell labeling: A potential radioactive agent for labeling human neutrophils. J Nucl Med 22:32, 1981. 7452353 [Google Scholar]
- 134.Fischman AJ, Pike MC, Kroon D, et al. : Imaging focal sites of bacterial infection in rats with indium-111-labeled chemotactic peptide analogs. J Nucl Med 32:483–491, 1991 [PubMed] [Google Scholar]
- 135.Fischman AJ, Rauh D, Solomon H, et al. : In vivo bioactivity and biodistribution of chemotactic peptide analogs in nonhuman primates. J Nucl Med 34:2130–2134, 1993 [PubMed] [Google Scholar]
- 136.Fischman AJ, Babich JW, Rubin RH: Infection imaging with technetium-99m-labeled chemotactic peptide analogs. Semin Nucl Med 24:154–168, 1994 [DOI] [PubMed] [Google Scholar]
- 137.Babich JW, Graham W, Barrow SA, et al. : Technetium-99m-labeled chemotactic peptides: Comparison with indium-111-labeled white blood cells for localizing acute bacterial infection in the rabbit. J Nucl Med 34:2176–2181, 1993 [PubMed] [Google Scholar]
- 138.Murphy PM: Chemokines and the molecular basis of cancer metastasis. N Engl J Med 345:833–835, 2001 [DOI] [PubMed] [Google Scholar]
- 139.Curfs JH, Meis JF, Hoogkamp-Korstanje JA: A primer on cytokines: Sources, receptors, effects, and inducers. Clin Microbiol Rev 10:742–780, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Fridman WH, Tartour E: The use of cytokines in the treatment of solid tumours. Hematol Cell Ther 39:105–108, 1997 [DOI] [PubMed] [Google Scholar]
- 141.Fridkin M, Najjar VA: Tuftsin: Its chemistry, biology, and clinical potential. Crit Rev Biochem Mol Biol 24:1–40, 1989 [DOI] [PubMed] [Google Scholar]
- 142.Peers SH, Tran LL, Eriksson SJ, et al. : Imaging a model of colitis with RP128, a Tc-99m-chelated tuftsin antagonist. J Nucl Med 36:114P, 1995, abstract [Google Scholar]
- 143.Iles DL, Goodbody AE, Cockeram A, et al. : A pilot phase II clinical study on imaging inflammatory lesions in patients with Crohn’s disease using Tc-99m RP128. J Nucl Med 39:271P, 1998, abstract [Google Scholar]
- 144.Caveliers V, Goodbody AE, Tran LL, et al. : Evaluation of 99mTc-RP128 as a potential inflammation imaging agent: Human dosimetry and first clinical results. J Nucl Med 42:154–161, 2001 [PubMed] [Google Scholar]
- 145.van der Laken CJ, Boerman OC, Oyen WJ, et al. : In-vivo expression of interleukin-1 receptors during various experimentally induced inflammatory conditions. J Infect Dis 177:1398–1401, 1998 [DOI] [PubMed] [Google Scholar]
- 146.Barrera P, van der Laken CJ, Boerman OC, et al. : Radiolabelled interleukin-1 receptor antagonist for detection of synovitis in patients with rheumatoid arthritis. Rheumatology (Oxford) 39:870–874, 2000 [DOI] [PubMed] [Google Scholar]
- 147.van der Laken CJ, Boerman OC, Oyen WJ, et al. : Imaging of infection in rabbits with radioiodinated interleukin-1 (alpha and beta), its receptor antagonist and a chemotactic peptide: A comparative study. Eur J Nucl Med 25:347–352, 1998 [DOI] [PubMed] [Google Scholar]
- 148.Chianelli M, Signore A, Fritzberg AR, et al. : The development of technetium-99m-labelled interleukin-2: A new radiopharmaceutical for the in vivo detection of mononuclear cell infiltrates in immune-mediated diseases. Nucl Med Biol 24:579–586, 1997 [DOI] [PubMed] [Google Scholar]
- 149.Signore A, Chianelli M, Annovazzi A, et al. : 123I-interleukin-2 scintigraphy for in vivo assessment of intestinal mononuclear cell infiltration in Crohn’s disease. J Nucl Med 41:242–249, 2000 [PubMed] [Google Scholar]
- 150.Annovazzi A, Biancone L, Caviglia R, et al. : 99mTc-interleukin-2 and (99m)Tc-HMPAO granulocyte scintigraphy in patients with inactive Crohn’s disease. Eur J Nucl Med Mol Imaging 30:374–382, 2003 [DOI] [PubMed] [Google Scholar]
- 151.Chianelli M, Mather SJ, Grossman A, et al. : 99mTc-interleukin-2 scintigraphy in normal subjects and in patients with autoimmune thyroid diseases: A feasibility study. Eur J Nucl Med Mol Imaging 35:2286–2293, 2008 [DOI] [PubMed] [Google Scholar]
- 152.Chianelli M, Parisella MG, Visalli N, et al. : Pancreatic scintigraphy with 99mTc-interleukin-2 at diagnosis of type 1 diabetes and after 1 year of nicotinamide therapy. Diabetes Metab Res Rev 24:115–122, 2008 [DOI] [PubMed] [Google Scholar]
- 153.Signore A, Picarelli A, Annovazzi A, et al. : 123I-Interleukin-2: Biochemical characterization and in vivo use for imaging autoimmune diseases. Nucl Med Commun 24:305–316, 2003 [DOI] [PubMed] [Google Scholar]
- 154.Signore A, Capriotti G, Chianelli M, et al. : Detection of insulitis by pancreatic scintigraphy with 99mTc-labeled IL-2 and MRI in patients with LADA (Action LADA 10). Diabetes Care 38:652–658, 2015 [DOI] [PubMed] [Google Scholar]
- 155.van der Laken CJ, Boerman OC, Oyen WJG, et al. : Targeting inflammation with radiolabelled interleukin-1 and other cytokines in various mouse models. Nucl Med Commun 18:478, 1997, abstract [Google Scholar]
- 156.Hoch RC, Schraufstätter IU, Cochrane CG: In vivo, in vitro, and molecular aspects of interleukin-8 and the interleukin-8 receptors. J Lab Clin Med 128:134–145, 1996 [DOI] [PubMed] [Google Scholar]
- 157.Luster AD: Chemokines—chemotactic cytokines that mediate inflammation. N Engl J Med 338:436–445, 1998 [DOI] [PubMed] [Google Scholar]
- 158.Hay RV, Skinner RS, Newman OC, et al. : Scintigraphy of acute inflammatory lesions in rats with radiolabelled recombinant human interleukin-8. Nucl Med Commun 18:367–378, 1997 [DOI] [PubMed] [Google Scholar]
- 159.Gross MD, Shapiro B, Fig LM, et al. : Imaging of human infection with (131)I-labeled recombinant human interleukin-8. J Nucl Med 42:1656–1659, 2001 [PubMed] [Google Scholar]
- 160.Rennen HJ, Boerman OC, Oyen WJ, et al. : Specifi and rapid scintigraphic detection of infection with 99mTc-labeled interleukin-8. J Nucl Med 42:117–123, 2001 [PubMed] [Google Scholar]
- 161.Bleeker-Rovers CP, Rennen HJ, Boerman OC, et al. : 99mTc-labeled interleukin 8 for the scintigraphic detection of infection and inflammation First clinical evaluation. J Nucl Med 48:337–343, 2007 [PubMed] [Google Scholar]
- 162.Scott P: IL-12: Initiation cytokine for cell-mediated immunity. Science 260:496–497, 1993 [DOI] [PubMed] [Google Scholar]
- 163.Stern AS, Magram J, Presky DH: Interleukin-12 an integral cytokine in the immune response. Life Sci 58:639–654, 1996 [DOI] [PubMed] [Google Scholar]
- 164.Annovazzi A, D’Alessandria C, Bonanno E, et al. : Synthesis of 99mTc-HYNIC-interleukin-12, a new specifi radiopharmaceutical for imaging T lymphocytes. Eur J Nucl Med Mol Imaging 33:474–482, 2006 [DOI] [PubMed] [Google Scholar]
- 165.Moyer BR, Vallabhajosula S, Lister-James J, et al. : Technetium-99m-white blood cell-specific imaging agent developed from platelet factor 4 to detect infection. J Nucl Med 37:673–679, 1996 [PubMed] [Google Scholar]
- 166.Palestro CJ, Weiland FL, Seabold JE, et al. : Localizing infection with a technetium-99m-labeled peptide: Initial results. Nucl Med Commun 22:695–701, 2001 [DOI] [PubMed] [Google Scholar]
- 167.Toda A, Yokomizo T, Shimizu T: Leukotriene B4 receptors. Prostaglandins Other Lipid Mediat 68-69:575–585, 2002 [DOI] [PubMed] [Google Scholar]
- 168.van Eerd JE, Oyen WJ, Harris TD, et al. : A bivalent leukotriene B(4) antagonist for scintigraphic imaging of infectious foci. J Nucl Med 44:1087–1091, 2003 [PubMed] [Google Scholar]
- 169.van Eerd JE, Oyen WJ, Harris TD, et al. : Scintigraphic imaging of infectious foci with an 111In-LTB4 antagonist is based on in vivo labeling of granulocytes. J Nucl Med 46:786–793, 2005 [PubMed] [Google Scholar]
- 170.Tulchinsky M, Peters AM: Leukocyte receptor-binding radiopharmaceuticals for infection and inflammation scintigraphy. J Nucl Med 46:718–721, 2005 [PubMed] [Google Scholar]
- 171.Méndez-Samperio P: Recent advances in the field of antimicrobial peptides in inflammatory diseases. Adv Biomed Res 2:50, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Epand RM, Vogel HJ: Diversity of antimicrobial peptides and their mechanisms of action. Biochim Biophys Acta 1462:11–28, 1999 [DOI] [PubMed] [Google Scholar]
- 173.Welling MM, Paulusma-Annema A, Balter HS, et al. : Technetium-99m labelled antimicrobial peptides discriminate between bacterial infections and sterile inflammations. Eur J Nucl Med 27:292–301, 2000 [DOI] [PubMed] [Google Scholar]
- 174.Chianelli M, Boerman OC, Malviya G, et al. : Receptor binding ligands to image infection. Curr Pharm Des 14:3316–3325, 2008 [DOI] [PubMed] [Google Scholar]
- 175.Lupetti A, Pauwels EK, Nibbering PH, et al. : 99mTc-antimicrobial peptides: Promising candidates for infection imaging. Q J Nucl Med 47:238–245, 2003 [PubMed] [Google Scholar]
- 176.Welling MM, Lupetti A, Balter HS, et al. : 99mTc-labeled antimicrobial peptides for detection of bacterial and Candida albicans infections. J Nucl Med 42:788–794, 2001 [PubMed] [Google Scholar]
- 177.Lichtenstein A: Mechanism of mammalian cell lysis mediated by peptide defensins. Evidence for an initial alteration of the plasma membrane. J Clin Invest 88:93–100, 1991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Levy O : Antibiotic proteins of polymorphonuclear leukocytes. Eur J Haematol 56:263–277, 1996 [DOI] [PubMed] [Google Scholar]
- 179.Welling MM, Nibbering PH, Paulusma-Annema A, et al. : Imaging of bacterial infections with 99mTc-labeled human neutrophil peptide-1. J Nucl Med 40:2073–2080, 1999 [PubMed] [Google Scholar]
- 180.Nibbering PH, Ravensbergen E, Welling MM, et al. : Human lactoferrin and peptides derived from its N terminus are highly effective against infections with antibiotic-resistant bacteria. Infect Immun 69:1469–1476, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Welling MM, Visentin R, Feitsma HI, et al. : Infection detection in mice using 99mTc-labeled HYNIC and N2S2 chelate conjugated to the antimicrobial peptide UBI 29–41. Nucl Med Biol 31:503–509, 2004 [DOI] [PubMed] [Google Scholar]
- 182.Welling MM, Mongera S, Lupetti A, et al. : Radiochemical and biological characteristics of 99mTc-UBI 29–41 for imaging of bacterial infections. Nucl Med Biol 29:413–422, 2002 [DOI] [PubMed] [Google Scholar]
- 183.Meléndez-Alafort L, Rodríguez-Cortés J, Ferro-Flores G, et al. : Biokinetics of (99m)Tc-UBI 29–41 in humans. Nucl Med Biol 31:373–379, 2004 [DOI] [PubMed] [Google Scholar]
- 184.Bhatt J, Mukherjee A, Korde A, et al. : Radiolabeling and preliminary evaluation of Ga-68 labeled NODAGA-ubiquicidin fragments for prospective infection imaging. Mol Imaging Biol 19:59–67, 2017 [DOI] [PubMed] [Google Scholar]
- 185.Rusckowski M, Gupta S, Liu G, et al. : Investigations of a (99m)Tc-labeled bacteriophage as a potential infection-specific imaging agent. J Nucl Med 45:1201–1208, 2004 [PubMed] [Google Scholar]
- 186.Rusckowski M, Gupta S, Liu G, et al. : Investigation of four (99m)Tc-labeled bacteriophages for infection-specific imaging. Nucl Med Biol 35:433–440, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Penn DL, Kim C, Zhang K, et al. : Apoptotic abscess imaging with 99mTc-HYNIC-rh-Annexin-V. Nucl Med Biol 37:29–34, 2010 [DOI] [PubMed] [Google Scholar]
- 188.Lorberboym M, Feldbrin Z, Hendel D, et al. : The use of 99mTc-recombinant human annexin V imaging for differential diagnosis of aseptic loosening and low-grade infection in hip and knee prostheses. J Nucl Med 50:534–537, 2009 [DOI] [PubMed] [Google Scholar]
- 189.Stadelmann C, Lassmann H: Detection of apoptosis in tissue sections. Cell Tissue Res 301:19–31, 2000 [DOI] [PubMed] [Google Scholar]
- 190.Tang C, Wang F, Hou Y, et al. : Technetium-99m-labeled annexin V imaging for detecting prosthetic joint infection in a rabbit model. J Biomed Res 29:224–231, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Thakur ML, Zhang K, Paudyal B, et al. : Targeting apoptosis for optical imaging of infection. Mol Imaging Biol 14:163–171, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Signore A, Mather SJ, Piaggio G, et al. : Molecular imaging of inflammation/infection: Nuclear medicine and optical imaging agents and methods. Chem Rev 110:3112–3145, 2010 [DOI] [PubMed] [Google Scholar]
- 193.Peterson KL, Reid WC, Freeman AF, et al. : The use of 14C-FIAU to predict bacterial thymidine kinase presence: Implications for radiolabeled FIAU bacterial imaging. Nucl Med Biol 40:638–642, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Bettegowda C, Foss CA, Cheong I, et al. : Imaging bacterial infections with radiolabeled 1-(2’-deoxy-2’-fluoro-beta-D-arabinofuranosyl)-5-iodouracil. Proc Natl Acad Sci USA 102:1145–1150, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Diaz LA Jr, Foss CA, Thornton K, et al. : Imaging of musculoskeletal bacterial infections by [124I]FIAU-PET/CT. PLoS ONE 2:e1007, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Zhang XM, Zhang HH, McLeroth P, et al. : [(124)I]FIAU: Human dosimetry and infection imaging in patients with suspected prosthetic joint infection. Nucl Med Biol 43:273–279, 2016 [DOI] [PubMed] [Google Scholar]
- 197.Ocakoglu K, Bayrak E, Onursal M, et al. : Evaluation of 99mTc-Pheophorbide-a use in infection imaging: A rat model. Appl Radiat Isot 69:1165–1168, 2011 [DOI] [PubMed] [Google Scholar]
- 198.Ning X, Seo W, Lee S, et al. : PET imaging of bacterial infections with fluorine maltohexaose. Angew Chem Int Ed Engl 53:14096–14101, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Gijs M, Aerts A, Impens N, et al. : Aptamers as radiopharmaceuticals for nuclear imaging and therapy. Nucl Med Biol 43:253–271, 2016 [DOI] [PubMed] [Google Scholar]
- 200.Hnatowich DJ: Pharmacokinetics of 99mTc-labeled oligonucleotides. Q J Nucl Med 40:202–208, 1996 [PubMed] [Google Scholar]
- 201.Chen L, Wang Y, Cheng D, et al. : (99m)Tc-MORF oligomers specific for bacterial ribosomal RNA as potential specific infection imaging agents. Bioorg Med Chem 21:6523–6530, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.dos Santos SR, Rodrigues Corra C, Branco de Barros AL, et al. : Identification of Staphylococcus aureus infection by aptamers directly radiolabeled with technetium-99m. Nucl Med Biol 42:292–298, 2015 [DOI] [PubMed] [Google Scholar]
- 203.de Sousa Lacerda CM, Ferreira IM, Dos Santos SR, et al. : (1→ 3)-β-D-glucan aptamers labeled with technetium-99m: Biodistribution and imaging in experimental models of bacterial and fungal infection. Nucl Med Biol 46:19–24, 2017 [DOI] [PubMed] [Google Scholar]
- 204.Petrik M, Franssen GM, Haas H, et al. : Preclinical evaluation of two 68Ga-siderophores as potential radiopharmaceuticals for Aspergillus fumigatus infection imaging. Eur J Nucl Med Mol Imaging 39:1175–1183, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Petrik M, Zhai C, Haas H, et al. : Siderophores for molecular imaging applications. Clin Transl Imaging 5:15–27, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]