Skip to main content
Erschienen in: EJNMMI Research 1/2014

Open Access 01.12.2014 | Original research

Could 18 F-DPA-714 PET imaging be interesting to use in the early post-stroke period?

verfasst von: Maria-Joao Ribeiro, Johnny Vercouillie, Severine Debiais, Jean-Philippe Cottier, Isabelle Bonnaud, Vincent Camus, Samuel Banister, Michael Kassiou, Nicolas Arlicot, Denis Guilloteau

Erschienen in: EJNMMI Research | Ausgabe 1/2014

Abstract

Background

Cerebral stroke is a severe and frequent condition that requires rapid and reliable diagnosis. If administered shortly after the first symptoms manifest themselves, IV thrombolysis has been shown to increase the functional prognosis by restoring brain reperfusion. However, a better understanding of the pathophysiology of stroke should help to identify potential new therapeutic targets. Stroke is known to induce an inflammatory brain reaction that involves overexpression of the 18-kDa translocator protein (TSPO) in glial cells and infiltrated leukocytes, which can be visualised by positron emission tomography (PET). We aimed to evaluate post-stroke neuroinflammation using the PET TSPO radioligand 18 F-DPA-714.

Methods

Nine patients underwent 18 F-DPA-714 PET and magnetic resonance imaging (MRI) between 8 and 18 days after the ictus. Co-registration of MRI and PET images was used to define three volumes of interest (VOIs): core infarction, contralateral region, and cerebellum ipsilateral to the stroke lesion. Time activity curves were obtained from each VOI, and ratios of mean and maximum activities between the VOIs were calculated.

Results

We observed an increased uptake of 18 F-DPA-714 co-localised with the infarct tissue and extension beyond the region corresponding to the damage in the blood brain barrier. No correlation was identified between 18 F-DPA-714 uptake and infarct volume. 18 F-DPA-714 uptake in ischemic lesion (mainly associated with TSPO expression in the infarct area and in the surrounding neighbourhood) slowly decreased from 10 min pi to the end of the PET acquisition, remaining higher than that in both contralateral region and ipsilateral cerebellum.

Conclusion

Our results show that 18 F-DPA-714 uptake after acute ischemia is mainly associated with TSPO expression in the infarct area and in the surrounding neighbourhood. We also demonstrated that the kinetics of 18 F-DPA-714 differs in injured tissue compared to normal tissue. Therefore, 18 F-DPA-714 may be useful in assessing the extent of neuroinflammation associated with acute stroke and could also help to predict clinical outcomes and functional recovery, as well as to assess therapeutic strategies, such as the use of neuroprotective/anti-inflammatory drugs.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13550-014-0028-4) contains supplementary material, which is available to authorized users.

Competing interests

All authors confirm that they have no applicable financial disclosures and no current or potential conflicts of interest.

Authors' contributions

MJR and JPC performed data acquisition and analysis. SB and MK performed the precursor production. JV, NA, and DG performed the radiosynthesis and pharmacological controls. SD and IB have recruited the patients and performed clinical examinations. NA performed statistical analysis. MJR, JV, JPC, VC, NA and DG drafted the manuscript. MJR, JPC, SD, IB, and DG conceived the study and participated in its design and coordination. All authors read and approved the final manuscript.

Background

Cerebral stroke is a severe and frequently occurring condition that represents a leading cause of mortality and morbidity worldwide [1, 2], being also the main aetiology of adult-acquired disabilities [3, 4]. Stroke is haemorrhagic in 10% to 15% of cases but more often it is ischemic (85% to 90%) [3]. Due to arterial occlusion, ischemic stroke is a diagnostic and therapeutic priority; emergency treatment (e.g., intravenous thrombolysis) should be administered in the first hours after symptom onset. Unfortunately, no other emergency drug treatment has been validated, but an improved understanding of the pathophysiology of early cerebral ischemia should identify potential molecular targets, particularly for neuroprotective treatments. Therefore, promising neuroprotective drugs, which demonstrated effectiveness in animals, have been evaluated; however, these results remain unconfirmed in clinical trials [5, 6]. Moreover, there is a lack of reliable imaging strategies to assess brain neuroinflammation after stroke [79].
Cerebral ischemia rapidly evolves to necrosis and a peri-necrosis area of ischemic penumbra, in which the brain tissue is still viable for a few hours. This cerebral tissue can be preserved if treatment is initiated quickly to restore the cerebral blood flow. Thus, this area of ‘darkness’ is the prime target for potential neuroprotective drugs. In animals, the infarcted area has been shown to expand in 24 h after the occlusion of a cerebral artery [10, 11].
Although different mechanisms are involved in the pathogenesis of stroke, increasing evidence suggests that inflammation, mainly involving the microglial and the immune system cells, account for its pathogenic progression [11].
Experimental studies have demonstrated that microglial cells are the first inflammatory cells activated after the onset of cerebral ischemia [12]. After breakdown of the blood-brain barrier (BBB), which accompanies cerebral ischemia, the perivascular microglia and macrophages are activated; this activation appears to be at least partially responsible for the inflammatory lesions in cerebral infarctions [12]. Microglia, which constitute up to 10% of the total brain cell population, change from a resting to an activated state in response to central nervous system insults, and this change stimulates these cells for phagocytosis. Several studies focusing on the inflammatory reaction during the first days after stroke have demonstrated that this inflammatory response changes dramatically over time [13, 14]. Therefore, the inflammatory markers correlated to the time course must be considered for any anti-inflammatory treatment approach in patients with acute ischemic strokes [14].
In humans, microglial activation can be assessed in vivo through neuroimaging of the 18-kDa translocator protein (TSPO) with selective TSPO radioligands. TSPO, formerly known as peripheral benzodiazepine receptor (PBR), is part of a multimeric ‘protein complex’ associated with the outer mitochondrial membrane of many cells [15]. TSPO is present in peripheral tissues and also in glial cells, but in the healthy brain, its expression is minimal [16]. TSPO may therefore be a valuable biomarker of inflammation, as it is highly expressed in phagocytic inflammatory cells.
A large number of positron emission tomography (PET) and single photon emission computerised tomography (SPECT) radioligands, selective for TSPO, have been developed, of which 11C-PK11195 was the first to be evaluated [17]. Studies using 11C-PK11195 demonstrated increased binding of this radiotracer around the outer border of ischemic lesions several days after stroke, as well as in distant areas from the lesion [1823]. This increased uptake was observed from 3 days after ictus, reaching its maximum at 7 days, and continuing for a period of 4 weeks [9].
However, the 20-min radioactive half-life of 11C-PK11195 is a serious drawback to the increased accessibility of biomarkers for routine clinical purposes since the use of these markers is limited to centres with an on-site cyclotron. Fluorine-18-labelled ligands, therefore, appear to be the best alternative, as the 110-min half-life of fluorine-18 enables centralised production and loco-regional delivery. Several groups have also evaluated neuroinflammation models in rats using the TSPO radioligand 18 F-DPA-714, concluding that it provides accurate quantitative information of TSPO density after cerebral ischemia, herpes encephalitis, amyotrophic lateral sclerosis, and gliomas [2427]. Several studies have already demonstrated that the regional distribution of 18 F-DPA-714 aligned well with other PET studies using TSPO radioligands in the human brain [25, 26].
The aim of the present study was to assess microglial activation after a recent stroke using 18 F-DPA-714, and to evaluate the relationship between 18 F-DPA-714 uptake and the infarct volume, also analysed by magnetic resonance imaging (MRI).

Methods

Subjects

This study was approved by the local Medical Bioethics Committee and was conducted according to French legislation and European directives.
Nine patients (Table 1) with recent unilateral cerebral infarcts were included. All patients underwent MRI and PET brain scans, with a maximum interval of 24 h between the two acquisitions. The following inclusion criteria were applied: Patients aged between 18 and 85 years, with a recent cerebral infarct involving the medial cerebral artery, visualised on computed tomography (CT) or MRI images, 6 to 20 days before the PET examination. None of the patients received the thrombolytic treatment, and none showed any symptoms suggesting the presence of any other significant neurodegenerative or psychiatric disease or had severe renal insufficiency, contraindicating the injection of a gadolinium chelate. No subject had been treated with anti-inflammatory drugs in the month preceding the PET study or with drugs that might interfere with 18 F-DPA-714 binding. All patients provided informed written consent before participating in the study.
Table 1
Clinical characteristics of the patients included in the study
Patient
Gender
Injury vascular region
Age (years)
Weight (kg)
Time between stroke and PET (days)
1
F
Right MCA
82
73
14
2
M
Left MCA
55
88
11
3
F
Right MCA
71
94
13
4
F
Left MCA
82
62
13
5
M
Right MCA
71
68
12
6
M
Right MCA
63
97
8
7
F
Right MCA
77
53
11
8
M
Left MCA
56
54
13
9
F
Right MCA
58
62
18
MCA, middle cerebral artery.

Radiosynthesis

N,N-diethyl-2-(2-(4-(2-fluoroethoxy)phenyl)-5,7-dimethyl-pyrazolo[1,5-α]pyrimidin-3-yl)acetamide (DPA-714) was labelled with fluorine-18 at its 2-fluoroethyl moiety, following nucleophilic substitution of the corresponding tosylate analogue, according to slight modifications of previously reported procedures [28]. The formulation of 18 F-DPA-714 provided a sterile injectable solution of isotonic sodium chloride with ethanol at a mass percentage of <8% for total injected volumes (ranging from 3 to 5 mL), in accordance with the European Pharmacopoeia. The mean specific activity of 18 F-DPA-714 obtained was 113.9 ± 36.9 GBq/μmol.

Imaging data acquisition

Brain MRI studies were performed using a 1.5 T imager (GE Healthcare, Milwaukee, WI, USA). Diffusion, FLAIR T2, T2*-weighted GRE sequences and high-resolution T1-weighted 3D MRI volumes were acquired for all subjects before and after the gadolinium administration (Gadobenate dimeglumine, Bracco imaging®, Ceriano Laghetto, Italy).
FLAIR T2-weighted and diffusion-weighted images showed the extent of cerebral infarction, whereas the T1-weighted SPGR image was used to allow co-registration with the PET images. After the gadolinium injection, axial volumetric T1 acquisition was performed to clarify the extent of the BBB breakdown.
PET studies were performed using a Dual Gemini (Philips Medical Systems, Amsterdam, the Netherlands), a whole-body hybrid PET/CT scanner with a full-width-half-maximum (FWHM) resolution of 5 mm in all directions, in 3D acquisition mode. To perform attenuation correction, a low dose CT helical scan was done (scan field, 600 mm; increments, 5 mm; slice thickness, 3.2 mm; pitch, 1.5, 0.75 second per rotation; 512 × 512 matrix; 120 kV; 80 mAs). Acquisition data were reconstructed with the standard package included with the system (PET view software-Philips Medical Systems). PET sinograms were corrected for tissue attenuation, decay, scatter, and random radiation, and then they were reconstructed using a 3D iterative RAMLA algorithm in voxels of 2 × 2 × 2 mm3.
To limit incidental movement, the patient's head was positioned in a headrest using a 3D laser alignment. All cerebral PET examinations were acquired in list mode over 90 min, after IV injection of 244.0 ± 27.4 MBq of 18 F-DPA-714.

Data analysis

An integrated PET image was obtained for each subject from the 90-min acquisition. This image was used to perform co-registration with each corresponding MRI image (T1-weighted SPGR and post-contrast images). Non-rigid registration was performed using normalised mutual information and PMOD® 3.4 software.
For each subject, a volume of interest (VOI) was defined over the injured tissue (IT) using individual MRI images. A similar VOI was transposed according to the region on the contralateral (CL) unaffected hemisphere. A VOI was also defined over the ipsilateral cerebellum to the injured tissue. All of VOIs were transposed onto PET images to obtain time activity curves (TACs). TACs were used to evaluate the kinetics of 18 F-DPA-714 over the 90-min acquisition. Ratios between the mean activity of the injured tissue VOIs and the contralateral (IT/CL) and cerebellar (IT/C) VOIs were calculated. We also determined similar ratios using the maximal activity measured in the same VOI.
For the nine patients, the infarct volumes (cm3) were calculated using the corresponding MRI images. Infarct volumes were manually outlined on diffusion-weighted MR images by an experienced neuroradiologist using an imaging analysis tool (functool, advantage windows, version 4.3; GE healthcare).

Statistical analysis

All data are presented as the mean ± standard deviation of the mean (SD). Significance was set at p < 0.05. Wilcoxon signed-rank test for paired values was used to determine the significant difference between VOIs ratios. Spearman's correlation test was used to assess the relationship between the volume of the lesion and 18 F-DPA-714 uptake ratios and between lesion volume measured by MRI and PET.

Results

The patients' clinical characteristics are summarised in Table 1. No adverse or subjective effects were observed for any of the nine subjects studied after injecting an average of 244.0 ± 27.4 MBq of 18 F-DPA-714. The imaging studies were performed 12.6 ± 2.7 days (min 8 days, max 18 days) after the acute injury.
Figure 1 shows an example of MRI and 18 F-DPA-714 images obtained from one patient (patient 4). For this patient, we observed that the radiopharmaceutical uptake is relatively comparable to injury tissue highlighted by MRI.
Figure 2 depicts a representative TAC of 18 F-DPA-714 uptake in the injured tissue and in the intact counterpart contralateral region and ipsilateral cerebellum (both used as reference regions) from a single subject. Figure 3 shows the evolution of the ratios calculated between the mean activity of the injured region and the two reference regions for the same subject. For all patients, the TAC revealed an initial rapid accumulation of radioactivity in the injured tissue, with a peak observed at 5 min post-injection (pi), followed by a gradual cerebral clearance from 10 to 20 min pi to the end of the PET acquisition. For the two reference regions, lower binding values of 18 F-DPA-714 were observed, with a maximum uptake at 5 min pi followed by two decreased phases from 5 to 20 min pi and from 10 to 20 min pi to the end of the acquisition. Moreover, the binding of 18 F-DPA-714 was lower for the contralateral region than for the cerebellum. For all subjects, the 18 F-DPA-714 uptake in the infarcted tissue remained higher than that observed for the other two regions during the entire acquisition.
Using the two reference regions at 40 and 80 min pi, the ratios obtained for the nine patients are presented in Table 2. The ratios using the mean activities were higher, although they were significantly different only at 40 min pi, when the contralateral VOI was used as the reference region, compared with the ratios using the cerebellum (IT/CL = 1.78 ± 0.31 and IT/Cerebellum = 1.55 ± 0.21 at 40 min, p = 0.020; IT/CL = 1.99 ± 0.56 and IT/Cerebellum = 1.79 ± 0.40 at 80 min pi, p = 0.155 respectively). Using the maximal activity calculated for each VOI, the ratios were also higher, however not significantly different, when using contralateral VOI (IT/CL = 2.05 ± 0.54 and IT/Cerebellum = 2.02 ± 0.64 at 40 min, p = 0.911; IT/CL = 2.44 ± 0.72 and IT/Cerebellum = 2.19 ± 0.54 at 80 min pi, p = 0.164, respectively).
Table 2
Volumes (cm 3 ) of infarcted region and VOIs and individual ratio values
Patient
Infarct volume
VOI volume
40 min post-injection
80 min post-injection
 
(cm3)
(cm3)
IT/CL
IT/Ipsilateral cerebellum
IT/CL
IT/Ipsilateral cerebellum
 
MRI
PET
Mean
Max
Mean
Max
Mean
Max
Mean
Max
1
108
93.0
2.03
2.75
1.70
2.70
2.03
2.85
1.77
2.47
2
16
6.0
1.14
1.08
1.26
1.21
1.28
1.27
1.29
1.30
3
190
163.8
2.09
2.11
159
1.82
3.27
3.42
2.55
2.59
4
13.1
17.3
2.10
2.61
1.80
2.89
2.05
2.32
1.77
2.36
5
34.6
32.3
1.93
2.14
1.55
2.54
2.08
3.41
1.64
2.62
6
3.4
3.5
1.54
1.84
1.17
1.47
1.46
1.67
1.25
1.39
7
0.7
0.73
1.83
1.68
1.73
2.52
2.08
2.39
1.82
2.82
8
12.9
13.7
1.73
2.54
1.67
1.60
1.69
2.52
2.07
2.23
9
1.6
1.8
1.66
1.67
1.48
1.42
1.95
2.15
1.96
1.89
Mean
  
1.78
2.05
1.55
2.02
1.99
2.44
1.79
2.19
SD
  
0.31
0.54
0.21
0.64
0.56
0.72
0.40
0.54
For each VOI, mean activity and maximal (Max) activity were used. The mean and standard deviation (SD) for these ratios are also indicated. Individual ratio values were calculated between the injured tissue (IT, necrotic and inflammatory) and the two reference regions, contralateral (CL) and ipsilateral cerebellum.
The median lesion volume estimated by MRI was 13.1 cm3 [range 0.7 (subject 7) to 190 cm3 (subject 3)]. There was no statistically significant relationship between this volume and the 18 F-DPA-714 uptake, according to Spearman's test (p = 0.24). However, a strong correlation was observed between the lesion volume estimated by MRI and the VOI volume used to calculate the activity concentration (r = 0.95 end p = 0.0003, Spearman's test).

Discussion

In this study, all imaging scans were performed within 12.6 ± 2.7 days after the ictus. Studies performed with the reference TSPO radioligand 11C-PK11195 in stroke patients indicated that the binding became significant a few days after the stroke onset, subsequently increased in approximately 1 week, and declined after 3 to 4 weeks [18, 19, 29]. A more recent study with 11C-PK11195, conducted 2 to 3 weeks post-stroke in a group of 18 subjects confirmed the local activation of microglia with increased radioligand uptake in the infarcted area [23]. Moreover, activation in the peri-infarct area appears to be more intense than in the core [30]. The value of our study is that it demonstrates the ability of a TSPO fluorine radioligand to map inflammatory process, in both the infarct and the surrounding surviving tissues. Indeed, one of the main difficulties in validating new radiopharmaceuticals as biomarkers of neuroinflammation lies in ruling out the possibility of non-specific binding, particularly passive leakage through a damaged BBB, as well as potential infiltration of peripheral circulating monocytes/macrophages within the brain parenchyma.
We observed that the increased uptake of 18 F-DPA-714 on PET images differed, even if very moderately, from the MRI gadolinium-enhanced regions, and was not limited to the infarct tissue with BBB breakdown; it also included non-infarcted cerebral tissue. This finding most likely reflects the activation of microglia with an increase in TSPO expression at the periphery of the necrotic lesion. On the other hand, we cannot forget that the PET spatial resolution is lower than that of the MRI's.
Moreover, the radiotracer uptake was not correlated with the infarct volume measured by MRI. This point is in accordance with a previous USPIO MRI study performed in subacute stroke patients demonstrating that the extent of the inflammatory response after stroke was unrelated to the lesion volume [31].
To our knowledge, only one study has compared two different TSPO radioligands in post-stroke patients (i.e., vinpocetine and PK11195, both labelled with carbon-11) [32]. The authors compared the diagnostic potential of these radioligands to visualise activated microglia in four post-stroke patients. They observed that the binding potential for 11C-vinpocetine was greater than that obtained for 11C-PK11195 in all evaluated regions, but the differences did not reach significance. 11C-vinpocetine binding potential was greater in the peri-infarcted zone than in the ischaemic core, but the difference did not prove to be significant [32, 33].
However, a direct comparison between 18 F-DPA-714 and 11C-PK11195 labelling, on a model of cerebral ischemia in rats, was done by Boutin et al. [34]. 18 F-DPA-714 displayed a higher signal-to-noise ratio than 11C-PK11195, suggesting that with the longer half-life of fluorine-18, 18 F-DPA-714 could be a good alternative for TSPO imaging.
The TACs generated for each subject showed that 18 F-DPA-714 uptake in the lesion, as well as in the contralateral cerebral tissue and the cerebellum, reached peak values at 5 min pi. The TACs for the injured region showed a slow decrease in binding from the maximum uptake to the end of the acquisition. However, this kinetic analysis showed two decreased phases for the other two regions: (1) a faster phase, between 5 and approximately 20 min, and (2) a slower phase, from 20 min pi to the end of the acquisition. This evolution of the kinetics of 18 F-DPA-714 in the cerebellum and in the normal cerebral tissue aligned with that observed in a previous study performed in a group of seven healthy volunteers [25]. Indeed, in this previous study, we observed that the kinetics of the radioligand was similar for all cortical regions and the cerebellum.
On the other hand, stroke patients may show remote metabolic changes in the cerebellum resulting from diaschisis [25, 35]. Although microglial changes, as a consequence of cerebellar diaschisis, are not known, we have chosen to take into account only the cerebellar hemisphere ipsilateral to the stroke to define the reference region.
The evolution of the 18 F-DPA-714 uptake in humans differed from that reported by Martin et al. in an ischemia model in rats [36]. The authors showed that the 18 F-DPA-714 uptake in the lesion reached a peak value at 30 min and remained stable until the end of the acquisition. In our study, we observed an earlier peak at 5 min pi followed by a slower decrease.
We also demonstrated that the binding ratio of 18 F-DPA-714 was higher when the contralateral VOI was used as a reference region, compared to when the cerebellum was used as a reference region. Several authors have previously documented the fact that the cerebellum in humans was not devoid of TSPO [37] and it would be of interest in future TSPO PET imaging studies to focus on focal inflammation, including stroke, Rasmussen encephalitis, epilepsy, and traumatic brain injury, and to consider the contralateral area as a suitable and TSPO-expression free reference tissue. Nevertheless, Martin et al. also detected increased 18 F-DPA-714 binding in the contralateral side at 11 days after focal cerebral ischemia in rats. They concluded that this finding was probably due to different mechanisms related to the expansion of infarction, such as spreading depression [36]. Thus, the exact fraction of specific binding to TSPO, in both the contralateral region and the cerebellum, would have to be determined by a blocking study requiring substantial doses of a blocking agent, which was ethically inconsistent with the present study. Interestingly, the ratio values calculated in the present study between the injured tissue and the contralateral region at 40 min post-18 F-DPA-714 injection, using the mean activity values, were significantly higher (p = 0.020, Mann-Whitney U test) than those previously published by Gerhard et al. using 11C-PK11195, obtained with three acquisitions from patients with the same specifications (interval in the range of 6 to 20 days between stroke ictus and PET acquisition) [18]. Although this comparison of two separate studies should be considered cautiously, it suggests that 18 F-DPA-714 might represent a good fluorine-18 alternative to 11C-PK11195 for TSPO PET imaging, with the higher ratio of ipsilateral-to-contralateral uptake. Moreover, our results are in accordance with preclinical direct comparison of both radiopharmaceuticals in a rodent model of focal ischemia [34], demonstrating that 18 F-DPA-714 achieved in vivo a higher ratio of ipsilateral uptake to contralateral uptake than 11C-PK11195.
Several studies have demonstrated the existence of different binder populations for 11C-PBR28, for which approximately 10% of the population appeared to be non-binders [38]. Owen and colleagues evidenced 3 types of binding pattern: high-affinity binders (approximately 50%), low-affinity binders (approximately 20%) and mixed-affinity binders (approximately 30%), related to a single-nucleotide polymorphism (rs6971) within the human TSPO gene [39], and they extended this finding to other PET TSPO radioligands, including the carbon 11-radiolabelled derivative of 18 F-DPA-714, namely, DPA-713 [40]. One limitation of our study is that we did not directly identify the subjects with low or high affinity for TSPO. It is likely that 18 F-DPA-714 is also sensitive to this variable inter-individual affinity state. Nevertheless, all patients in our study demonstrated increased 18 F-DPA-714 uptake by the injured tissue.

Conclusions

Our results show that the uptake of 18 F-DPA-714 occurred not only due to the BBB breakdown, but also because of the activation of microglia in the area surrounding the infarct. We also showed that the kinetics of 18 F-DPA-714 differ between injured and normal tissues. The ability to assess microglial activation in vivo may improve our understanding of the mechanisms of neuroinflammation in acute disorders such as stroke, and should enable effective treatment monitoring. This study also demonstrates that a 10 to 20 min of PET emission acquisition, performed between 40 and 80 min pi, was able to differentiate acute tissue injury from normal brain tissue.

Acknowledgements

We thank Dr Marie Gaudron-Assor and the technical and nursing staffs of CERRP, the Service de Médecine Nucléaire, INSERM CIC 1415, INSERM CIC 1415-IT, and CHRU Tours for their assistance. This work was partially supported by grants from CHRU Tours (Grant NCT 00563537), Radex and COSMI Programs get partially financial supports from the European Union (Europe is engaged in Région Centre with the FEDER, Fonds européen de développement régional), the French National Agency for Research (‘Investissements d’Avenir’ n°ANR-11-LABX-0018-01, IRON), and Laboratoires Cyclopharma. The research leading to these results has received funding from the European Union's Seventh Framework Programme (FP7/2007-2013) under grant agreement n° 278850 (INMiND).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 2.0 International License (https://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

All authors confirm that they have no applicable financial disclosures and no current or potential conflicts of interest.

Authors' contributions

MJR and JPC performed data acquisition and analysis. SB and MK performed the precursor production. JV, NA, and DG performed the radiosynthesis and pharmacological controls. SD and IB have recruited the patients and performed clinical examinations. NA performed statistical analysis. MJR, JV, JPC, VC, NA and DG drafted the manuscript. MJR, JPC, SD, IB, and DG conceived the study and participated in its design and coordination. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Bikbov B, Perico N, Remuzzi G: Mortality landscape in the global burden of diseases, injuries and risk factors study. Eur J Intern Med 2014, 25: 1–5. 10.1016/j.ejim.2013.09.002CrossRefPubMed Bikbov B, Perico N, Remuzzi G: Mortality landscape in the global burden of diseases, injuries and risk factors study. Eur J Intern Med 2014, 25: 1–5. 10.1016/j.ejim.2013.09.002CrossRefPubMed
2.
Zurück zum Zitat The state of US health, 1990–2010: burden of diseases, injuries, and risk factors JAMA 2013, 310: 591–608. 10.1001/jama.2013.13805 The state of US health, 1990–2010: burden of diseases, injuries, and risk factors JAMA 2013, 310: 591–608. 10.1001/jama.2013.13805
3.
Zurück zum Zitat Donnan GA, Fisher M, Macleod M, Davis SM: Stroke. Lancet 2008, 371: 1612–1623. 10.1016/S0140-6736(08)60694-7CrossRefPubMed Donnan GA, Fisher M, Macleod M, Davis SM: Stroke. Lancet 2008, 371: 1612–1623. 10.1016/S0140-6736(08)60694-7CrossRefPubMed
4.
Zurück zum Zitat Lo EH, Dalkara T, Moskowitz MA: Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci 2003, 4: 399–415. 10.1038/nrn1106CrossRefPubMed Lo EH, Dalkara T, Moskowitz MA: Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci 2003, 4: 399–415. 10.1038/nrn1106CrossRefPubMed
5.
Zurück zum Zitat Lai AY, Todd KG: Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. Glia 2006, 53: 809–816. 10.1002/glia.20335CrossRefPubMed Lai AY, Todd KG: Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. Glia 2006, 53: 809–816. 10.1002/glia.20335CrossRefPubMed
6.
Zurück zum Zitat Tymianski M: Novel approaches to neuroprotection trials in acute ischemic stroke. Stroke 2013, 44: 2942–2950. 10.1161/STROKEAHA.113.000731CrossRefPubMed Tymianski M: Novel approaches to neuroprotection trials in acute ischemic stroke. Stroke 2013, 44: 2942–2950. 10.1161/STROKEAHA.113.000731CrossRefPubMed
7.
Zurück zum Zitat Deddens LH, Van Tilborg GA, Mulder WJ, De Vries HE, Dijkhuizen RM: Imaging neuroinflammation after stroke: current status of cellular and molecular MRI strategies. Cerebrovasc Dis 2012, 33: 392–402. 10.1159/000336116CrossRefPubMed Deddens LH, Van Tilborg GA, Mulder WJ, De Vries HE, Dijkhuizen RM: Imaging neuroinflammation after stroke: current status of cellular and molecular MRI strategies. Cerebrovasc Dis 2012, 33: 392–402. 10.1159/000336116CrossRefPubMed
8.
9.
Zurück zum Zitat Thiel A, Heiss WD: Imaging of microglia activation in stroke. Stroke 2011, 42: 507–512. 10.1161/STROKEAHA.110.598821CrossRefPubMed Thiel A, Heiss WD: Imaging of microglia activation in stroke. Stroke 2011, 42: 507–512. 10.1161/STROKEAHA.110.598821CrossRefPubMed
10.
Zurück zum Zitat del Zoppo GJ, Pessin MS, Mori E, Hacke W: Thrombolytic intervention in acute thrombotic and embolic stroke. Semin Neurol 1991, 11: 368–384. 10.1055/s-2008-1041241CrossRefPubMed del Zoppo GJ, Pessin MS, Mori E, Hacke W: Thrombolytic intervention in acute thrombotic and embolic stroke. Semin Neurol 1991, 11: 368–384. 10.1055/s-2008-1041241CrossRefPubMed
11.
Zurück zum Zitat Mabuchi T, Kitagawa K, Ohtsuki T, Kuwabara K, Yagita Y, Yanagihara T, Hori M, Matsumoto M: Contribution of microglia/macrophages to expansion of infarction and response of oligodendrocytes after focal cerebral ischemia in rats. Stroke 2000, 31: 1735–1743. 10.1161/01.STR.31.7.1735CrossRefPubMed Mabuchi T, Kitagawa K, Ohtsuki T, Kuwabara K, Yagita Y, Yanagihara T, Hori M, Matsumoto M: Contribution of microglia/macrophages to expansion of infarction and response of oligodendrocytes after focal cerebral ischemia in rats. Stroke 2000, 31: 1735–1743. 10.1161/01.STR.31.7.1735CrossRefPubMed
12.
Zurück zum Zitat Danton GH, Dietrich WD: Inflammatory mechanisms after ischemia and stroke. J Neuropathol Exp Neurol 2003, 62: 127–136.PubMed Danton GH, Dietrich WD: Inflammatory mechanisms after ischemia and stroke. J Neuropathol Exp Neurol 2003, 62: 127–136.PubMed
13.
Zurück zum Zitat Rallidis LS, Vikelis M, Panagiotakos DB, Rizos I, Zolindaki MG, Kaliva K, Kremastinos DT: Inflammatory markers and in-hospital mortality in acute ischaemic stroke. Atherosclerosis 2006, 189: 193–197. 10.1016/j.atherosclerosis.2005.11.032CrossRefPubMed Rallidis LS, Vikelis M, Panagiotakos DB, Rizos I, Zolindaki MG, Kaliva K, Kremastinos DT: Inflammatory markers and in-hospital mortality in acute ischaemic stroke. Atherosclerosis 2006, 189: 193–197. 10.1016/j.atherosclerosis.2005.11.032CrossRefPubMed
14.
Zurück zum Zitat Worthmann H, Tryc AB, Deb M, Goldbecker A, Ma YT, Tountopoulou A, Lichtinghagen R, Weissenborn K: Linking infection and inflammation in acute ischemic stroke. Ann N Y Acad Sci 2010, 1207: 116–122. 10.1111/j.1749-6632.2010.05738.xCrossRefPubMed Worthmann H, Tryc AB, Deb M, Goldbecker A, Ma YT, Tountopoulou A, Lichtinghagen R, Weissenborn K: Linking infection and inflammation in acute ischemic stroke. Ann N Y Acad Sci 2010, 1207: 116–122. 10.1111/j.1749-6632.2010.05738.xCrossRefPubMed
15.
Zurück zum Zitat Papadopoulos V, Baraldi M, Guilarte TR, Knudsen TB, Lacapère JJ, Lindemann P, Norenberg MD, Nutt D, Weizman A, Zhang MR, Gavish M: Translocator protein (18 kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci 2006, 27: 402–409. 10.1016/j.tips.2006.06.005CrossRefPubMed Papadopoulos V, Baraldi M, Guilarte TR, Knudsen TB, Lacapère JJ, Lindemann P, Norenberg MD, Nutt D, Weizman A, Zhang MR, Gavish M: Translocator protein (18 kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci 2006, 27: 402–409. 10.1016/j.tips.2006.06.005CrossRefPubMed
16.
Zurück zum Zitat Veenman L, Gavish M: Peripheral-type benzodiazepine receptors: their implication in brain disease. Drug Develop Res 2000, 50: 355–370. 10.1002/1098-2299(200007/08)50:3/4<355::AID-DDR18>3.0.CO;2-WCrossRef Veenman L, Gavish M: Peripheral-type benzodiazepine receptors: their implication in brain disease. Drug Develop Res 2000, 50: 355–370. 10.1002/1098-2299(200007/08)50:3/4<355::AID-DDR18>3.0.CO;2-WCrossRef
17.
Zurück zum Zitat Petit-Taboue MC, Baron JC, Barre L, Travère JM, Speckel D, Camsonne R, MacKenzie ET: Brain kinetics and specific binding of [11C]PK11195 to omega 3 sites in baboons: positron emission tomography study. Eur J Pharmacol 1991, 200: 347–351. 10.1016/0014-2999(91)90594-GCrossRefPubMed Petit-Taboue MC, Baron JC, Barre L, Travère JM, Speckel D, Camsonne R, MacKenzie ET: Brain kinetics and specific binding of [11C]PK11195 to omega 3 sites in baboons: positron emission tomography study. Eur J Pharmacol 1991, 200: 347–351. 10.1016/0014-2999(91)90594-GCrossRefPubMed
18.
Zurück zum Zitat Gerhard A, Neumaier B, Elitok E, Glatting G, Ries V, Tomczak R, Ludolph AC, Reske SN: In vivo imaging of activated microglia using [11C]PK11195 and positron emission tomography in patients after ischemic stroke. Neuroreport 2000, 11: 2957–2960. 10.1097/00001756-200009110-00025CrossRefPubMed Gerhard A, Neumaier B, Elitok E, Glatting G, Ries V, Tomczak R, Ludolph AC, Reske SN: In vivo imaging of activated microglia using [11C]PK11195 and positron emission tomography in patients after ischemic stroke. Neuroreport 2000, 11: 2957–2960. 10.1097/00001756-200009110-00025CrossRefPubMed
19.
Zurück zum Zitat Gerhard A, Schwarz J, Myers R, Wise R, Banati RB: Evolution of microglial activation in patients after ischemic stroke: a [11C](R)-PK11195 PET study. Neuroimage 2005, 24: 591–595. 10.1016/j.neuroimage.2004.09.034CrossRefPubMed Gerhard A, Schwarz J, Myers R, Wise R, Banati RB: Evolution of microglial activation in patients after ischemic stroke: a [11C](R)-PK11195 PET study. Neuroimage 2005, 24: 591–595. 10.1016/j.neuroimage.2004.09.034CrossRefPubMed
20.
Zurück zum Zitat Radlinska BA, Ghinani SA, Lyon P, Jolly D, Soucy JP, Minuk J, Schirrmacher R, Thiel A: Multimodal microglia imaging of fiber tracts in acute subcortical stroke. Ann Neurol 2009, 66: 825–832. 10.1002/ana.21796CrossRefPubMed Radlinska BA, Ghinani SA, Lyon P, Jolly D, Soucy JP, Minuk J, Schirrmacher R, Thiel A: Multimodal microglia imaging of fiber tracts in acute subcortical stroke. Ann Neurol 2009, 66: 825–832. 10.1002/ana.21796CrossRefPubMed
21.
Zurück zum Zitat Rojas S, Martin A, Arranz MJ, Pareto D, Purroy J, Verdaguer E, Llop J, Gómez V, Gispert JD, Millán O, Chamorro A, Planas AM: Imaging brain inflammation with [11C]PK11195 by PET and induction of the peripheral-type benzodiazepine receptor after transient focal ischemia in rats. J Cereb Blood Flow Metab 2007, 27: 1975–1986. 10.1038/sj.jcbfm.9600500CrossRefPubMed Rojas S, Martin A, Arranz MJ, Pareto D, Purroy J, Verdaguer E, Llop J, Gómez V, Gispert JD, Millán O, Chamorro A, Planas AM: Imaging brain inflammation with [11C]PK11195 by PET and induction of the peripheral-type benzodiazepine receptor after transient focal ischemia in rats. J Cereb Blood Flow Metab 2007, 27: 1975–1986. 10.1038/sj.jcbfm.9600500CrossRefPubMed
22.
Zurück zum Zitat Schroeter M, Dennin MA, Walberer M, Backes H, Neumaier B, Fink GR, Graf R: Neuroinflammation extends brain tissue at risk to vital peri-infarct tissue: a double tracer [11C]PK11195- and [18 F]FDG-PET study. J Cereb Blood Flow Metab 2009, 29: 1216–1225. 10.1038/jcbfm.2009.36CrossRefPubMed Schroeter M, Dennin MA, Walberer M, Backes H, Neumaier B, Fink GR, Graf R: Neuroinflammation extends brain tissue at risk to vital peri-infarct tissue: a double tracer [11C]PK11195- and [18 F]FDG-PET study. J Cereb Blood Flow Metab 2009, 29: 1216–1225. 10.1038/jcbfm.2009.36CrossRefPubMed
23.
Zurück zum Zitat Thiel A, Radlinska BA, Paquette C, Sidel M, Soucy JP, Schirrmacher R, Minuk J: The temporal dynamics of poststroke neuroinflammation: a longitudinal diffusion tensor imaging-guided PET study with 11C-PK11195 in acute subcortical stroke. J Nucl Med 2010, 51: 1404–1412. 10.2967/jnumed.110.076612CrossRefPubMed Thiel A, Radlinska BA, Paquette C, Sidel M, Soucy JP, Schirrmacher R, Minuk J: The temporal dynamics of poststroke neuroinflammation: a longitudinal diffusion tensor imaging-guided PET study with 11C-PK11195 in acute subcortical stroke. J Nucl Med 2010, 51: 1404–1412. 10.2967/jnumed.110.076612CrossRefPubMed
24.
Zurück zum Zitat Abourbeh G, Theze B, Maroy R, Dubois A, Brulon V, Fontyn Y, Dollé F, Tavitian B, Boisgard R: Imaging microglial/macrophage activation in spinal cords of experimental autoimmune encephalomyelitis rats by positron emission tomography using the mitochondrial 18 kDa translocator protein radioligand [18 F]DPA-714. J Neurosci 2012, 32: 5728–5736. 10.1523/JNEUROSCI.2900-11.2012CrossRefPubMed Abourbeh G, Theze B, Maroy R, Dubois A, Brulon V, Fontyn Y, Dollé F, Tavitian B, Boisgard R: Imaging microglial/macrophage activation in spinal cords of experimental autoimmune encephalomyelitis rats by positron emission tomography using the mitochondrial 18 kDa translocator protein radioligand [18 F]DPA-714. J Neurosci 2012, 32: 5728–5736. 10.1523/JNEUROSCI.2900-11.2012CrossRefPubMed
25.
Zurück zum Zitat Arlicot N, Vercoullie J, Ribeiro MJ, Tauber C, Venel Y, Baulieu JL, Maia S, Corcia P, Stabin MG, Reynolds A, Kassiou M, Guilloteau D: Initial evaluation in healthy humans of [18 F]DPA-714, a potential PET biomarker for neuroinflammation. Nucl Med Biol 2012, 39: 570–578. 10.1016/j.nucmedbio.2011.10.012CrossRefPubMed Arlicot N, Vercoullie J, Ribeiro MJ, Tauber C, Venel Y, Baulieu JL, Maia S, Corcia P, Stabin MG, Reynolds A, Kassiou M, Guilloteau D: Initial evaluation in healthy humans of [18 F]DPA-714, a potential PET biomarker for neuroinflammation. Nucl Med Biol 2012, 39: 570–578. 10.1016/j.nucmedbio.2011.10.012CrossRefPubMed
26.
Zurück zum Zitat Corcia P, Tauber C, Vercoullie J, Arlicot N, Prunier C, Praline J, Nicolas G, Venel Y, Hommet C, Baulieu JL, Cottier JP, Roussel C, Kassiou M, Guilloteau D, Ribeiro MJ: Molecular imaging of microglial activation in amyotrophic lateral sclerosis. PLoS One 2012, 7: e52941. 10.1371/journal.pone.0052941PubMedCentralCrossRefPubMed Corcia P, Tauber C, Vercoullie J, Arlicot N, Prunier C, Praline J, Nicolas G, Venel Y, Hommet C, Baulieu JL, Cottier JP, Roussel C, Kassiou M, Guilloteau D, Ribeiro MJ: Molecular imaging of microglial activation in amyotrophic lateral sclerosis. PLoS One 2012, 7: e52941. 10.1371/journal.pone.0052941PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Tang D, Hight MR, McKinley ET, Fu A, Buck JR, Smith RA, Tantawy MN, Peterson TE, Colvin DC, Ansari MS, Nickels M, Manning HC: Quantitative preclinical imaging of TSPO expression in glioma using N, N-diethyl-2-(2-(4-(2–18 F-fluoroethoxy)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide. J Nucl Med 2012, 53: 287–294. 10.2967/jnumed.111.095653PubMedCentralCrossRefPubMed Tang D, Hight MR, McKinley ET, Fu A, Buck JR, Smith RA, Tantawy MN, Peterson TE, Colvin DC, Ansari MS, Nickels M, Manning HC: Quantitative preclinical imaging of TSPO expression in glioma using N, N-diethyl-2-(2-(4-(2–18 F-fluoroethoxy)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide. J Nucl Med 2012, 53: 287–294. 10.2967/jnumed.111.095653PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat James ML, Fulton RR, Vercoullie J, Henderson DJ, Garreau L, Chalon S, Dolle F, Costa B, Guilloteau D, Kassiou M: DPA-714, a new translocator protein-specific ligand: synthesis, radiofluorination, and pharmacologic characterization. J Nucl Med 2008, 49: 814–822. 10.2967/jnumed.107.046151CrossRefPubMed James ML, Fulton RR, Vercoullie J, Henderson DJ, Garreau L, Chalon S, Dolle F, Costa B, Guilloteau D, Kassiou M: DPA-714, a new translocator protein-specific ligand: synthesis, radiofluorination, and pharmacologic characterization. J Nucl Med 2008, 49: 814–822. 10.2967/jnumed.107.046151CrossRefPubMed
29.
Zurück zum Zitat Pappata S, Levasseur M, Gunn RN, Myers R, Crouzel C, Syrota A, Jones T, Kreutzberg GW, Banati RB: Thalamic microglial activation in ischemic stroke detected in vivo by PET and [11C]PK1195. Neurology 2000, 55: 1052–1054. 10.1212/WNL.55.7.1052CrossRefPubMed Pappata S, Levasseur M, Gunn RN, Myers R, Crouzel C, Syrota A, Jones T, Kreutzberg GW, Banati RB: Thalamic microglial activation in ischemic stroke detected in vivo by PET and [11C]PK1195. Neurology 2000, 55: 1052–1054. 10.1212/WNL.55.7.1052CrossRefPubMed
30.
Zurück zum Zitat Price CJ, Wang D, Menon DK, Guadagno JV, Cleij M, Fryer T, Aigbirhio F, Baron JC, Warburton EA: Intrinsic activated microglia map to the peri-infarct zone in the subacute phase of ischemic stroke. Stroke 2006, 37: 1749–1753. 10.1161/01.STR.0000226980.95389.0bCrossRefPubMed Price CJ, Wang D, Menon DK, Guadagno JV, Cleij M, Fryer T, Aigbirhio F, Baron JC, Warburton EA: Intrinsic activated microglia map to the peri-infarct zone in the subacute phase of ischemic stroke. Stroke 2006, 37: 1749–1753. 10.1161/01.STR.0000226980.95389.0bCrossRefPubMed
31.
Zurück zum Zitat Nighoghossian N, Wiart M, Cakmak S, Berthezène Y, Derex L, Cho TH, Nemoz C, Chapuis F, Tisserand GL, Pialat JB, Trouillas P, Froment JC, Hermier M: Inflammatory response after ischemic stroke: a USPIO-enhanced MRI study in patients. Stroke 2007, 38: 303–307. 10.1161/01.STR.0000254548.30258.f2CrossRefPubMed Nighoghossian N, Wiart M, Cakmak S, Berthezène Y, Derex L, Cho TH, Nemoz C, Chapuis F, Tisserand GL, Pialat JB, Trouillas P, Froment JC, Hermier M: Inflammatory response after ischemic stroke: a USPIO-enhanced MRI study in patients. Stroke 2007, 38: 303–307. 10.1161/01.STR.0000254548.30258.f2CrossRefPubMed
32.
Zurück zum Zitat Gulyas B, Toth M, Vas A, Shchukin E, Kostulas K, Hillert J, Halldin C: Visualising neuroinflammation in post-stroke patients: a comparative PET study with the TSPO molecular imaging biomarkers [11C]PK11195 and [11C]vinpocetine. Curr Radiopharm 2012, 5: 19–28. 10.2174/1874471011205010019CrossRefPubMed Gulyas B, Toth M, Vas A, Shchukin E, Kostulas K, Hillert J, Halldin C: Visualising neuroinflammation in post-stroke patients: a comparative PET study with the TSPO molecular imaging biomarkers [11C]PK11195 and [11C]vinpocetine. Curr Radiopharm 2012, 5: 19–28. 10.2174/1874471011205010019CrossRefPubMed
33.
Zurück zum Zitat Gulyas B, Toth M, Schain M, Airaksinen A, Vas A, Kostulas K, Lindström P, Hillert J, Halldin C: Evolution of microglial activation in ischaemic core and peri-infarct regions after stroke: a PET study with the TSPO molecular imaging biomarker [11C]vinpocetine. J Neurol Sci 2012, 320: 110–117. 10.1016/j.jns.2012.06.026CrossRefPubMed Gulyas B, Toth M, Schain M, Airaksinen A, Vas A, Kostulas K, Lindström P, Hillert J, Halldin C: Evolution of microglial activation in ischaemic core and peri-infarct regions after stroke: a PET study with the TSPO molecular imaging biomarker [11C]vinpocetine. J Neurol Sci 2012, 320: 110–117. 10.1016/j.jns.2012.06.026CrossRefPubMed
34.
Zurück zum Zitat Boutin H, Prenant C, Maroy R, Galea J, Greenhalgh AD, Smigova A, Cawthorne C, Julyan P, Wilkinson SM, Banister SD, Brown G, Herholz K, Kassiou M, Rothwell NJ: [18 F]DPA-714: direct comparison with [11C]PK11195 in a model of cerebral ischemia in rats. PLoS One 2013, 8: e56441. 10.1371/journal.pone.0056441PubMedCentralCrossRefPubMed Boutin H, Prenant C, Maroy R, Galea J, Greenhalgh AD, Smigova A, Cawthorne C, Julyan P, Wilkinson SM, Banister SD, Brown G, Herholz K, Kassiou M, Rothwell NJ: [18 F]DPA-714: direct comparison with [11C]PK11195 in a model of cerebral ischemia in rats. PLoS One 2013, 8: e56441. 10.1371/journal.pone.0056441PubMedCentralCrossRefPubMed
35.
36.
Zurück zum Zitat Martin A, Boisgard R, Theze B, Van Camp N, Kuhnast B, Damont A, Kassiou M, Dollé F, Tavitian B: Evaluation of the PBR/TSPO radioligand [18 F]DPA-714 in a rat model of focal cerebral ischemia. J Cereb Blood Flow Metab 2010, 30: 230–241. 10.1038/jcbfm.2009.205PubMedCentralCrossRefPubMed Martin A, Boisgard R, Theze B, Van Camp N, Kuhnast B, Damont A, Kassiou M, Dollé F, Tavitian B: Evaluation of the PBR/TSPO radioligand [18 F]DPA-714 in a rat model of focal cerebral ischemia. J Cereb Blood Flow Metab 2010, 30: 230–241. 10.1038/jcbfm.2009.205PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Gavish M, Bachman I, Shoukrun R, Katz Y, Veenman L, Weisinger G, Weizman A: Enigma of the peripheral benzodiazepine receptor. Pharmacol Rev 1999, 51: 629–650.PubMed Gavish M, Bachman I, Shoukrun R, Katz Y, Veenman L, Weisinger G, Weizman A: Enigma of the peripheral benzodiazepine receptor. Pharmacol Rev 1999, 51: 629–650.PubMed
38.
Zurück zum Zitat Kreisl WC, Fujita M, Fujimura Y, Kimura N, Jenko KJ, Kannan P, Hong J, Morse CL, Zoghbi SS, Gladding RL, Jacobson S, Oh U, Pike VW, Innis RB: Comparison of [11C]-(R)-PK 11195 and [11C]PBR28, two radioligands for translocator protein (18 kDa) in human and monkey: implications for positron emission tomographic imaging of this inflammation biomarker. Neuroimage 2010, 49: 2924–2932. 10.1016/j.neuroimage.2009.11.056PubMedCentralCrossRefPubMed Kreisl WC, Fujita M, Fujimura Y, Kimura N, Jenko KJ, Kannan P, Hong J, Morse CL, Zoghbi SS, Gladding RL, Jacobson S, Oh U, Pike VW, Innis RB: Comparison of [11C]-(R)-PK 11195 and [11C]PBR28, two radioligands for translocator protein (18 kDa) in human and monkey: implications for positron emission tomographic imaging of this inflammation biomarker. Neuroimage 2010, 49: 2924–2932. 10.1016/j.neuroimage.2009.11.056PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Owen DR, Yeo AJ, Gunn RN, Song K, Wadsworth G, Lewis A, Rhodes C, Pulford DJ, Bennacef I, Parker CA, StJean PL, Cardon LR, Mooser VE, Matthews PM, Rabiner EA, Rubio JP: An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28. J Cereb Blood Flow Metab 2012, 32: 1–5. 10.1038/jcbfm.2011.147PubMedCentralCrossRefPubMed Owen DR, Yeo AJ, Gunn RN, Song K, Wadsworth G, Lewis A, Rhodes C, Pulford DJ, Bennacef I, Parker CA, StJean PL, Cardon LR, Mooser VE, Matthews PM, Rabiner EA, Rubio JP: An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28. J Cereb Blood Flow Metab 2012, 32: 1–5. 10.1038/jcbfm.2011.147PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Owen DR, Gunn RN, Rabiner EA, Bennacef I, Fujita M, Kreisl WC, Innis RB, Pike VW, Reynolds R, Matthews PM, Parker CA: Mixed-affinity binding in humans with 18-kDa translocator protein ligands. J Nucl Med 2011, 52: 24–32. 10.2967/jnumed.110.079459PubMedCentralCrossRefPubMed Owen DR, Gunn RN, Rabiner EA, Bennacef I, Fujita M, Kreisl WC, Innis RB, Pike VW, Reynolds R, Matthews PM, Parker CA: Mixed-affinity binding in humans with 18-kDa translocator protein ligands. J Nucl Med 2011, 52: 24–32. 10.2967/jnumed.110.079459PubMedCentralCrossRefPubMed
Metadaten
Titel
Could 18 F-DPA-714 PET imaging be interesting to use in the early post-stroke period?
verfasst von
Maria-Joao Ribeiro
Johnny Vercouillie
Severine Debiais
Jean-Philippe Cottier
Isabelle Bonnaud
Vincent Camus
Samuel Banister
Michael Kassiou
Nicolas Arlicot
Denis Guilloteau
Publikationsdatum
01.12.2014
Verlag
Springer Berlin Heidelberg
Erschienen in
EJNMMI Research / Ausgabe 1/2014
Elektronische ISSN: 2191-219X
DOI
https://doi.org/10.1186/s13550-014-0028-4

Weitere Artikel der Ausgabe 1/2014

EJNMMI Research 1/2014 Zur Ausgabe