Skip to main content
Erschienen in: EJNMMI Research 1/2018

Open Access 01.12.2018 | Original research

PET imaging of 68Ga-NODAGA-RGD, as compared with 18F-fluorodeoxyglucose, in experimental rodent models of engrafted glioblastoma

verfasst von: Sibel Isal, Julien Pierson, Laetitia Imbert, Alexandra Clement, Charlotte Collet, Sophie Pinel, Nicolas Veran, Aurélie Reinhard, Sylvain Poussier, Guillaume Gauchotte, Steeven Frezier, Gilles Karcher, Pierre-Yves Marie, Fatiha Maskali

Erschienen in: EJNMMI Research | Ausgabe 1/2018

Abstract

Background

Tracers triggering αvβ3 integrins, such as certain RGD-containing peptides, were found promising in previous pilot studies characterizing high-grade gliomas. However, only limited comparisons have been performed with current PET tracers. This study aimed at comparing the biodistribution of 18F-fluorodeoxyglucose (18F-FDG) with that of 68Ga-NODAGA-RGD, an easily synthesized monomeric RGD compound with rapid kinetics, in two different rodent models of engrafted human glioblastoma.

Methods

Nude rodents bearing human U87-MG glioblastoma tumor xenografts in the flank (34 tumors in mice) or in the brain (5 tumors in rats) were analyzed. Kinetics of 68Ga-NODAGA-RGD and of 18F-FDG were compared with PET imaging in the same animals, along with additional autohistoradiographic analyses and blocking tests for 68Ga-NODAGA-RGD.

Results

Both tracers showed a primary renal route of clearance, although with faster clearance for 68Ga-NODAGA-RGD resulting in higher activities in the kidneys and bladder. The tumor activity from 68Ga-NODAGA-RGD, likely corresponding to true integrin binding (i.e., suppressed by co-injection of a saturating excess of unlabeled RGD), was found relatively high, but only at the 2nd hour following injection, corresponding on average to 53% of total tumor activity. Tumor uptake of 68Ga-NODAGA-RGD decreased progressively with time, contrary to that of 18F-FDG, although 68Ga-NODAGA-RGD exhibited 3.4 and 3.7-fold higher tumor-to-normal brain ratios on average compared to 18F-FDG in mice and rat models, respectively. Finally, ex-vivo analyses revealed that the tumor areas with high 68Ga-NODAGA-RGD uptake also exhibited the highest rates of cell proliferation and αv integrin expression, irrespective of cell density.

Conclusions

68Ga-NODAGA-RGD has a high potential for PET imaging of glioblastomas, especially for areas with high integrin expression and cell proliferation, although PET recording needs to be delayed until the 2nd hour following injection in order to provide sufficiently high integrin specificity.

Background

High-grade gliomas constitute the most frequent cause of brain primary tumors and remain associated with a very poor prognosis under treatment [1]. Imaging plays a key role in their diagnosis and medical monitoring. Although magnetic resonance imaging is the first-line technique, positron emission tomography (PET) is increasingly used in this setting [2]. The diagnostic information obtained with 18F-fluorodeoxyglucose (18F-FDG) is somewhat variable whereas amino acid PET tracers, such as 11C-methionine, 18F-FDOPA, and 18F-FET, allow a more accurate, tumor characterization [3], which may also be the case for more recent radiotracers developed for PET imaging of αvβ3 integrins [4, 5].
These integrins are currently expressed in glioma cells and in neo-vessel endothelial cells, in conjunction with tumor-related angiogenic processes [6]. The expression of αvβ3 integrins increases with the malignancy grade of the glioma and appears particularly important in high-grade gliomas where αvβ3 integrins may facilitate tumor progression [7]. Most of the radiotracers that bind these integrins contain one or several tripeptide Arg-Gly-Asp (RGD) amino acid sequences owing to their high affinity for αvβ3 integrins [4, 5, 811]. While some of these RGD-containing peptides have been labeled with Fluor-18, their synthesis appears to be too complex for routine applications despite their high diagnostic potential [9, 12].
Other RGD-containing peptides, labeled with 68Ga through a metal complexation process, are much easier to synthesize [5, 10, 11]. This is notably the case of cyclic RGD peptides conjugated with a macrocyclic chelator, such as the dimeric 68Ga-NOTA-PRGD2 peptide, which was successfully tested in human gliomas and exhibiting a superiority to 18F-FDG in differentiating high- from low-grade gliomas [5]. However, such comparison with 18F-FDG has never been undertaken to date for 68Ga-NODAGA-RGD, a monomeric peptide containing only a single cyclic RGD (c(RGDyK)) and involving NODAGA as the chelating group [13]. Although the affinity for αvβ3 integrins is lower for monomeric than for multimeric RGD-containing tracers, this is seemingly counterbalanced by faster kinetics and diffusion rates, owing to their much smaller size and leading to a favorable biodistribution for tumor imaging, as shown in a previous pre-clinical study [10].
In light of the above, the present study aimed at comparing the biodistribution of 18F-FDG to that of 68Ga-NODAGA-RGD, a monomeric RGD-containing tracer with ease of synthesis and rapid kinetics, in two different rodent models of engrafted human glioblastoma.

Methods

Production and quality control of 68Ga-NODAGA-RGD

NODAGA-RGD was purchased from ABX GmbH (Hamburg, Germany). The 68Ga-labeling of NODAGA-RGD was performed using a mAIO synthesizer (Trasis, Ans, Belgium) with removable cassettes. The 68Ge/68Ga generator (IGG-100; Eckert & Ziegler Europe, 370 MBq) was eluted with 5 mL of a 0.1 M HCl solution. The eluate was used without pre-purification. NODAGA-RGD (10 μg, 10 nmol) was dissolved at 30 °C for 5 min in 1 mL of 0.7 M NaOAc buffer. The resulting product, 68Ga-NODAGA-RGD, was purified on a SepPak® C18 light cartridge, eluted with 50% (0.5/0.5 v/v) ethanol and completed with a 0.9% saline solution in order to reach a total volume of 5 mL. High-performance liquid chromatography (HPLC) analyses were performed on a Waters system (Milford, Massachusetts, USA) equipped with a 2695eb pump, an auto sampler injector, a UV spectrometer (2998 PDA detector), and a Berthold LB-500 radioactivity detector, controlled by the Empower software.
HPLC conditions consisted of an ACE C18 column (AIT, 150 × 3 mm, 3 μm), flow rate 0.5 mL/min at 25 °C, acetonitrile (CH3CN)/H2O/0.1% trifluoroacetic acid (TFA) gradient, 0–2 min 0% CH3CN, and 2–16 min 0–40% CH3CN.

Animal models and experimental plan

All protocols were approved by the Lorraine Ethics Committee N°68 according to Guidelines of Animal Care and Use (APAFIS no. 2456-2015102609499994).
Tumor xenografts were obtained in mice flanks as described previously [14]. Six-week-old female immunodeficient (nu/nu) NMRI mice, weighing 20–25 g, were obtained from Janvier Laboratories (Le Genest Saint Isle, France) and housed in ventilated cages including filter tops with an ad libitum access to food and water. Two xenograft tumors were induced in all mice through subcutaneous flank injections of 2.106 of human U87-MG cells. Tumor growth was assessed twice weekly, according to tumor volume (in mm3) calculated with the formula d2 × D/2, where d and D represent the shortest and longest diameters, respectively, calculated in millimeter with a caliper (decrescent PRC, EDVC, France).
Seventeen of these mice, with tumor volumes ranging from 150 to 750 mm3, were referred to the experimental protocol involving two consecutive PET recording procedures at a 48-h interval, one performed after the injection of 68Ga-NODAGA-RGD and the other after the injection of 18F-FDG. In order to prevent any order effect, the 68Ga-NODAGA-RGD PET was performed first in half of the animals and second (after 18F-FDG injection) in the other half. At the end of the 2nd PET recording, the animals were sacrificed by inhalation of 5% of isoflurane followed by cervical dislocation. Tumors were subsequently surgically removed for further histopathological analysis.
The integrin specificity of 68Ga-NODAGA-RGD was assessed in supplementary blocking studies using in vivo PET imaging in five xenograft mice.
Finally, an additional comparison of the PET images provided by 18F-FDG and 68Ga-NODAGA-RGD was obtained in five rats, after brain stereotactic xenografts of the U87-MG cells, as described previously [15]. Briefly, 8-week-old athymic male nude rats (180–220 g), (Hsd: RH-Foxn1rnu; Envigo, Gannat, France), were anesthetized with a mixture of air and 1.5–2% isoflurane and were placed in a Kopf stereotactic frame (900 M Kopf Instruments, Tujunga, CA). After a scalp incision, a burr hole was drilled 0.5 mm anterior and 2.7 mm lateral to bregma and a skull anchor (Patent N° 11 55596) was affixed. Thereafter, approximately 5.104 U87-MG cells, suspended in 5 μL Hank’s Buffered Salt Solution (HBSS, 1×), were injected into the brain parenchyma at 4.4 mm depth. 18F-FDG and 68Ga-NODAGA-RGD PET were recorded 48-h apart, and 11 to 14 days after xenograft implantation.

Small animal PET studies

PET recordings were obtained with a camera dedicated to small animal studies (Inveon, Siemens Preclinical Solutions, Knoxville, USA).
Fasting mice were anesthetized with 1.5% isoflurane after which either 6 MBq of 68Ga-NODAGA-RGD or 15 MBq of 18F-FDG were injected as a bolus via a lateral tail vein cannulation after determination of baseline glycemia by a dedicated system (Optium Xceed, Abbott, France). List-mode acquisitions of 120 min durations were initiated a few seconds prior to tracer injection, and the acquired PET data were subsequently reconstructed in 27 consecutive frames (i.e., 5 frames of 120-s duration followed by 22 frames of 5-min duration) using the ordered-subsets expectation maximization 3D algorithm (OSEM3D, 4 iterations, 16 subsets, zoom 1) together with scatter and attenuation corrections based on transmission source measurement. The final voxel size was 0.8 × 0.8 × 0.9 mm3.
Tumor and of various organ activities were determined through SUV mean values within 3D regions of interests (ROI), which were drawn with a dedicated software (Inveon Research Workplace 4.1, Siemens®, Knoxville, USA) on the fusion images encompassing the entire 120-min recording period. Spheroid ROIs were placed inside the liver, brain, and kidney regions with the ROI limits approximating the organ limits as close as possible. Tumor and heart ROIs were obtained with isocontour ROIs and threshold limits of 50 and 80%, respectively, of the maximal voxel value. In this manner, no difference in ROI volumes was documented between 68Ga-NODAGA-RGD and 18F-FDG.
For the blocking studies performed in five additional xenograft mice, a native PET was recorded with 68Ga-NODAGA-RGD (5.9 ± 0.30 MBq) as described above, and this protocol was repeated unchanged 2 days later, except that 68Ga-NODAGA-RGD was co-injected with a saturating dose of unlabeled RGD (26 ± 3 mg kg−1 of c(RGDyK)). The tracer uptake was calculated through SUVmean values within the aforementioned described ROIs.
The 68Ga-NODAGA-RGD and 18F-FDG PET study protocols were unchanged for the rat experiments, aside from (i) higher activities were injected (approximately 40 MBq for 68Ga-NODAGA-RGD and 74 MBq of 18F-FDG), (ii) the PET recording was focused on the head area, and (iii) tumor activities, determined during the 2nd hour following tracer injection, were expressed relative to the activities from the contralateral normal brain (i.e., with the tumor ROI being duplicated and then moved to the contralateral area).

Gamma counter-based biodistribution study

Biodistribution was additionally analyzed ex vivo in tumor-bearing mice and for both 68Ga-NODAGA-RGD and 18F-FDG. Seventeen fasting mice were anesthetized with 1.5% isoflurane after which, 6 MBq of 68Ga-NODAGA-RGD (n = 8) or 15 MBq of 18F-FDG (n = 9) was injected as a bolus via a lateral tail vein. Animals were sacrificed by cervical dislocation 60 min after injection. Various tissues (tumor, liver, heart, brain, and kidney) were dissected, weighed, and counted in a calibrated gamma counter (Wizard, PerkinElmer, France). The results were expressed as percentages of injected dose and per gram of tissue (% ID/g).

Tissue sampling, autoradiography, and histology

Immediately after the last PET imaging protocol, the anesthetized animals were sacrificed by cervical dislocation. Tumors were subsequently removed surgically, and tissue samples were frozen in isopentane mixed with dry ice and sectioned in serial 20 and 8 μm sections for autohistoradiography and immunohistochemical staining, respectively. The distribution of 68Ga-NODAGA-RGD activity in the tumor sections was recorded with an autohistoradiography system dedicated to the detection of electrons and positrons (μImager™, Biospace, France). For further histopathological examination, adjacent tumor sections were fixed with paraformaldehyde 4%, stained with hematoxylin and eosin or with a specific rabbit polyclonal antibody to determine the expression of human αv integrin (anti-CD51, Cloud-Clone Corporation, PAB282Hu01) and Ki-67 protein (Thermo Fischer SP6 MAS-14520). Prior to incubation with antibodies, the tumor sections were incubated with a citrate buffer (pH 6.0) for 35 min at 97 °C.

Statistical analyses

All data are expressed as mean ± SEM. Statistical analyses were performed with SPSS Statistics Software v. 20 (IBM, NY, USA). Paired and unpaired comparisons of quantitative variables were performed with Student’s t tests and ANOVA tests, respectively. P values less than 0.05 were considered as statistically significant.

Results

Radiochemistry

An automated 68Ga-labeling of NODAGA-RGD resulted in a high labeling efficiency (> 98%) with a specific activity ranging from 20 to 30 MBq/nmol. The overall process, including synthesizer test, cassette test, radiosynthesis, and quality control leading to the production of around 250 MBq of 68Ga-NODAGA-RGD, lasted approximately 60 min. Radiochemical purity determined by HPLC was always > 95%.

PET imaging and biodistribution studies

The time-activity-curves (TACs), obtained in vivo for the tumors and for different organs after the injection of 68Ga-NODAGA-RGD or 18F-FDG on whole-body PET images of engrafted mice, are shown in Fig. 1. Both 68Ga-NODAGA-RGD and 18F-FDG showed a primary renal route of tracer clearance, although this clearance was faster for 68Ga-NODAGA-RGD resulting in particularly high activities in the kidneys and bladder. Tumor uptake of 68Ga-NODAGA-RGD decreased progressively with time, contrary to that of 18F-FDG for which a progressive enhancement was documented throughout the 2 h of analysis.
At 60 min post-injection, the mean tumor activity of 68Ga-NODAGA-RGD (SUVmean 0.23 ± 0.04) was one-fifth of that documented for 18F-FDG (1.19 ± 0.33; p < 0.01). By contrast, the TAC levels of the liver were comparable for the two tracers, and, as expected, heart and brain TAC levels were much higher for 18F-FDG (Fig. 1).
As detailed in Fig. 2, the tumor activity from 68Ga-NODAGA-RGD, which likely corresponded to true integrin binding (i.e., suppressed by the co-injection of a saturating excess of unlabeled RGD), was found relatively high, although only at the 2nd hour following injection, corresponding on average to 53% of the total tumor activity. By contrast, this saturation-related decrease was much lower (around 25%, on average), when computed in the 1st hour of PET recording.
As additionally shown in Fig. 2, a significant saturation-related decrease in 68Ga-NODAGA-RGD uptake was also observed on most of the other analyzed organs (liver, brain, and kidneys), although the amplitudes of these decreases were much lower than that observed for the tumor.
As detailed in Fig. 3, activity ratios between the tumor and various organs were computed for the second hour of PET recording and gamma counting, as well as in a comparative manner between 68Ga-NODAGA-RGD and 18F-FDG. These ratios were found lower with 68Ga-NODAGA-RGD than with 18F-FDG for tumor-to-liver but higher for tumor-to-heart or tumor-to-brain ratios. On average, with PET imaging, the tumor-to-normal brain ratio was 3.4 times higher for 68Ga-NODAGA-RGD (5.1 ± 1.4) than for 18F-FDG (1.5 ± 0.37) (Fig. 3a). This ratio was furthermore higher when computed with ex vivo biodistribution data (19.8 ± 10 vs. 0.61 ± 0.2%ID/g) for 68Ga-NODAGA-RGD and 18F-FDG, respectively (Fig. 3b).
Whole-body images of the 2nd hour of PET recordings are depicted in Fig. 4 for both tracers, providing an illustration of the potential of 68Ga-NODAGA-RGD for tumor imaging of thoracic and head areas, with much lower normal uptakes observed for 68Ga-NODAGA-RGD than for 18F-FDG in these areas.
A further illustration of the higher potential of 68Ga-NODAGA-RGD for the imaging of brain glioblastomas is provided in Fig. 5, from PET images recorded during the 2nd hour in a rat with an engrafted brain tumor. On average, in the five nude rats with engrafted glioblastomas, tumor activity expressed relative to that of the contralateral normal brain was found to be 3.73-fold higher for 68Ga-NODAGA-RGD (6.05 ± 0.67) than for 18F-FDG (1.62 ± 0.15) (see individual data in the Additional file 1: Table S1).

Ex vivo analyses

A representative example of the ex vivo analyses obtained after the injection of 68Ga-NODAGA-RGD is given in Fig. 6. Autohistoradiographic analyses with the μImager™ revealed a heterogeneous intratumoral distribution of 68Ga-NODAGA-RGD; of note, areas exhibiting a higher 68Ga-NODAGA-RGD activity were also those showing higher expressions of the integrin αv subunit and of the protein Ki-67, an indicator of cell proliferation. By contrast, in hematoxylin-eosin stained tissue slices, cells and nuclear densities were comparable between tumor areas with and without high 68Ga-NODAGA-RGD uptake (Fig. 6).

Discussion

In this experimental study, the kinetics and biodistribution of 68Ga-NODAGA-RGD, a monomeric RGD-containing tracer, were found to be more favorable for the PET imaging of glioblastoma than those of 18F-FDG, a reference tracer in oncology. Much higher tumor-to-normal brain ratios were indeed observed in two distinct rodent models of engrafted human U87-MG cells, one with tumors engrafted in mouse flanks and the other with direct engraftment in the rat brains.
The rationale behind these observations is that αvβ3 integrins, which are targeted by RGD-containing tracers, are highly expressed in high-grade gliomas in which the magnitude of this expression has been associated with the potential for tumor invasion, angiogenesis, and metastasis [6, 7, 1619]. Furthermore, these integrins are known to be poorly expressed within the different tissue components of normal brain and, in addition, the RGD-containing tracers may pass through the blood brain barrier when disrupted by a pathological process [8, 1822]. These considerations likely explain the high tumor-to-normal brain ratios documented herein for 68Ga-NODAGA-RGD, i.e., approximately five for the mouse model and six for the rat model, on average. This ratio was conversely much lower for 18F-FDG, i.e., approximately 1.5 for the mouse model and 1.6 for the rat model on average, owing to the variable glucose metabolism of high-grade gliomas [23] in addition to the high residual glucose metabolism of normal brain cells.
In further in vivo PET experiments, a seemingly high level of integrin binding of 68Ga-NODAGA-RGD within the tumors was documented by blocking experiments (i.e., with inhibition of integrin binding in the presence of saturating doses of unlabeled RGD). It should be pointed out, however, that this proportion of integrin binding was sufficiently high, i.e., corresponding to more than 50% of tumor activity measured on PET images, only during the second hour of the PET recording. This observation is in accordance with previous literature on RGD-containing tracers, leading to the current recommendation of delaying PET recording for 30 to 60 min after tracer injection [3, 10, 11]. This allows avoiding an early high level of nonspecific activity, possibly due to the circulating blood tracers, and leading to overestimate integrin density.
A significant saturation-related decrease in 68Ga-NODAGA-RGD uptake was also observed on most of the other analyzed organs, including the brain, although the amplitude of this decrease was much lower than for the tumor. Similar observations have also been reported for other RGD-containing radiotracers in both humans and animals [2428], owing to the fact that integrin αvβ3 is also expressed in various normal cells, including vascular smooth muscle cells, macrophages, and osteoclasts.
Further immunohistological analysis indicated a rather good correspondence between tumor sites, showing the highest level of 68Ga-NODAGA-RGD activity as well as high levels of αv integrin expression (Fig. 6), confirming the integrin specificity of this tracer. In these ex-vivo analyses, the heterogeneous tumor level of 68Ga-NODAGA-RGD activity was found to have a greater correspondence with the local distribution of cell proliferation (Ki-67 protein expression) rather than that of cell density. These observations further strengthen the general consideration that RGD-based radiotracers would be particularly indicated for characterizing high-grade gliomas showing high potential for tumor invasion, angiogenesis, and metastasis, as stated above.
On the other hand, these characteristics are likely to differ for 18F-FDG which rather reflects the distribution of glucose uptake and subsequent phosphorylation by the tumor cells. This may explain the lack of correlation noted herein between tumor uptake of 18F-FDG and that of 68Ga-NODAGA-RGD (results not shown), in accordance with a previous comparison performed with 18F-galacto-RGD in men with various cancer types [9, 29, 30].
18F-galacto-RGD is another monomeric RGD tracer, for which tumor uptake has been shown to be closely related to αvβ3 integrin expression, and was the first reported RGD-based tracer in human subjects [9, 31]. Unfortunately, the 18F-labeling of this tracer and of all RGD-containing peptides appears to be too complex for routine application [12]. RGD-containing peptides, labeled with 68Ga by a current metal complexation process, are conversely much easier to, synthesize.
This is particularly the case for our automated 68Ga-labeling process of NODAGA-RGD, resulting in a > 98% labeling efficiency and > 95% radiochemical purity. In a more general manner, the generator-produced 68Ga represents an attractive alternative to cyclotron-produced radionuclides in spite of a slightly lower image quality than that provided by 18F (owing to a higher energy of the emitted positron).
Our small monomeric RGD-containing tracer exhibited relatively rapid kinetics with a primary renal route of tracer clearance, which was faster than that of 18F-FDG, resulting in higher activities in the kidneys and bladder. It is likely that these somewhat high activities documented in the kidneys, bladder, and to a lesser extent in the liver may hamper imaging of abdominal tumors, as previously observed for other RGD-containing radiotracers [3, 10], although would likely not constitute a significant limitation for brain tumor imaging.
The fast-renal clearance of 68Ga-NODAGA-RGD was associated however, with a progressive decline in tumor activity with time, contrary to the progressive increase documented for 18F-FDG, which may constitute a limitation for brain imaging of high-grade gliomas. Taken together, as a result of this rather fast tumor clearance and the constraint of delaying PET recording to the 2nd hour following injection, dedicated clinical studies are thus mandatory to accurately ascertain the required level of activity to be injected for the late imaging of a sufficiently high tumor activity.
Additional comparisons are also required with amino acid PET tracers, such as 11C-methionin, 18F-FDOPA, and 18F-FET, as well as with dimeric RGD-containing tracers, which could provide an enhanced affinity for αvβ3 integrins though a slower diffusion rate when compared with monomeric forms [5].

Conclusions

This study confirms that 68Ga-NODAGA-RGD is a RGD compound with a high ease of synthesis and relatively rapid kinetics. Furthermore, experiments performed in two different rodent models of engrafted glioblastoma give evidence of a high potential for PET imaging, with very high tumor-to-normal brain activity ratios and a triggering of tumor areas with high integrin expression and high cell proliferation rates. However, it is likely that PET recording needs to be delayed until the 2nd hour following injection in order to yield sufficiently specific information on integrin density.

Acknowledgements

The authors thank Pierre Pothier, for critical review of the manuscript, and the Nancyclotep experimental imaging platform, for organizational support.
The authors thank Professor Simon Thornton for help with the English language.

Funding

The study was sponsored by the Nancy CHRU (research fellow grant) and supported by a grant from NANCYCLOTEP and Lorraine University (Nancyclotep call for proposal).

Availability of data and materials

Please contact author for data requests.

Ethics approval

All protocols for animal experimentation were performed in accordance with the international, national, and institutional guidelines for the care and use of animals, with the approval of the Lorraine Ethics Committee (CELMEA N°68) and under the current ethical regulations of the national law on animal experimentations (APAFIS no. 2456-2015102609499994).

Competing interests

The authors declare that they have no competing of interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
2.
Zurück zum Zitat Frosina G. Positron emission tomography of high-grade gliomas. J Neurooncol Mai. 2016;127(3):415–25.CrossRef Frosina G. Positron emission tomography of high-grade gliomas. J Neurooncol Mai. 2016;127(3):415–25.CrossRef
3.
Zurück zum Zitat Chen W. Clinical applications of PET in brain tumors. J Nucl Med. 2007;48(9):1468–81. Epub 2007 Aug 17.CrossRefPubMed Chen W. Clinical applications of PET in brain tumors. J Nucl Med. 2007;48(9):1468–81. Epub 2007 Aug 17.CrossRefPubMed
4.
Zurück zum Zitat Frosina G. Non-routine tracers for PET imaging of high-grade glioma. Anticancer Res. 2016;36(7):3253–60.PubMed Frosina G. Non-routine tracers for PET imaging of high-grade glioma. Anticancer Res. 2016;36(7):3253–60.PubMed
5.
Zurück zum Zitat Li D, Zhao X, Zhang L, Li F, Ji N, Gao Z, et al. (68)Ga-PRGD2 PET/CT in the evaluation of glioma: a prospective study. Mol Pharm. 2014;11(11):3923–9.CrossRefPubMedPubMedCentral Li D, Zhao X, Zhang L, Li F, Ji N, Gao Z, et al. (68)Ga-PRGD2 PET/CT in the evaluation of glioma: a prospective study. Mol Pharm. 2014;11(11):3923–9.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Schnell O, Krebs B, Wagner E, Romagna A, Beer AJ, Grau SJ, Thon N, Goetz C, Kretzschmar HA, Tonn JC, Goldbrunner RH. Expression of integrin alphavbeta3 in gliomas correlates with tumor grade and is not restricted to tumor vasculature. Brain Pathol. 2008;18(3):378–86.CrossRefPubMedPubMedCentral Schnell O, Krebs B, Wagner E, Romagna A, Beer AJ, Grau SJ, Thon N, Goetz C, Kretzschmar HA, Tonn JC, Goldbrunner RH. Expression of integrin alphavbeta3 in gliomas correlates with tumor grade and is not restricted to tumor vasculature. Brain Pathol. 2008;18(3):378–86.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Holmes KM, Annala M, Chua CY, Dunlap SM, Liu Y, Hugen N, Moore LM, Cogdell D, Hu L, Nykter M, Hess K, Fuller GN, Zhang W. Insulin-like growth factor-binding protein 2-driven glioma progression is prevented by blocking a clinically significant integrin, integrin-linked kinase, and NF-κB network. Proc Natl Acad Sci U S A. 2012;109(9):3475–80.CrossRefPubMedPubMedCentral Holmes KM, Annala M, Chua CY, Dunlap SM, Liu Y, Hugen N, Moore LM, Cogdell D, Hu L, Nykter M, Hess K, Fuller GN, Zhang W. Insulin-like growth factor-binding protein 2-driven glioma progression is prevented by blocking a clinically significant integrin, integrin-linked kinase, and NF-κB network. Proc Natl Acad Sci U S A. 2012;109(9):3475–80.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Nieberler M, Reuning U, Reichart F, Notni J, Wester HJ, Schwaiger M, Weinmüller M, Räder A, Steiger K, Kessler H. Exploring the role of RGD-recognizing Integrins in cancer. Cancers (Basel). 2017;9(9). Nieberler M, Reuning U, Reichart F, Notni J, Wester HJ, Schwaiger M, Weinmüller M, Räder A, Steiger K, Kessler H. Exploring the role of RGD-recognizing Integrins in cancer. Cancers (Basel). 2017;9(9). 
9.
Zurück zum Zitat Haubner R, Weber WA, Beer AJ, Vabuliene E, Reim D, Sarbia M, Becker KF, Goebel M, Hein R, Wester HJ, Kessler H, Schwaiger M. Noninvasive visualization of the activated alphavbeta3 integrin in cancer patients by positron emission tomography and [18F]Galacto-RGD. PLoS Med. 2005;2(3):e70. Epub 2005 Mar 29.CrossRefPubMedPubMedCentral Haubner R, Weber WA, Beer AJ, Vabuliene E, Reim D, Sarbia M, Becker KF, Goebel M, Hein R, Wester HJ, Kessler H, Schwaiger M. Noninvasive visualization of the activated alphavbeta3 integrin in cancer patients by positron emission tomography and [18F]Galacto-RGD. PLoS Med. 2005;2(3):e70. Epub 2005 Mar 29.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Li ZB, Chen K, Chen X. (68)Ga-labeled multimeric RGD peptides for microPET imaging of integrin alpha(v)beta (3) expression. Eur J Nucl Med Mol Imaging. 2008;35(6):1100–8.CrossRefPubMed Li ZB, Chen K, Chen X. (68)Ga-labeled multimeric RGD peptides for microPET imaging of integrin alpha(v)beta (3) expression. Eur J Nucl Med Mol Imaging. 2008;35(6):1100–8.CrossRefPubMed
11.
Zurück zum Zitat Knetsch PA, Petrik M, Griessinger CM, Rangger C, Fani M, Kesenheimer C, et al. (68Ga)NODAGA-RGD for imaging αvβ3 integrin expression. Eur J Nucl Med Mol Imaging. 2011;38(7):1303–12.CrossRefPubMed Knetsch PA, Petrik M, Griessinger CM, Rangger C, Fani M, Kesenheimer C, et al. (68Ga)NODAGA-RGD for imaging αvβ3 integrin expression. Eur J Nucl Med Mol Imaging. 2011;38(7):1303–12.CrossRefPubMed
12.
Zurück zum Zitat Haubner R, Kuhnast B, Mang C, Weber WA, Kessler H, Wester HJ, Schwaiger M. [18F]Galacto-RGD: synthesis, radiolabeling, metabolic stability, and radiation dose estimates. Bioconjug Chem. 2004;15(1):61–9.CrossRefPubMed Haubner R, Kuhnast B, Mang C, Weber WA, Kessler H, Wester HJ, Schwaiger M. [18F]Galacto-RGD: synthesis, radiolabeling, metabolic stability, and radiation dose estimates. Bioconjug Chem. 2004;15(1):61–9.CrossRefPubMed
13.
Zurück zum Zitat Pohle K, Notni J, Bussemer J, Kessler H, Schwaiger M, Beer AJ. 68Ga-NODAGA-RGD is a suitable substitute for (18)F-Galacto-RGD and can be produced with high specific activity in a cGMP/GRP compliant automated process. Nucl Med Biol. 2012;39(6):777–84.CrossRefPubMed Pohle K, Notni J, Bussemer J, Kessler H, Schwaiger M, Beer AJ. 68Ga-NODAGA-RGD is a suitable substitute for (18)F-Galacto-RGD and can be produced with high specific activity in a cGMP/GRP compliant automated process. Nucl Med Biol. 2012;39(6):777–84.CrossRefPubMed
14.
Zurück zum Zitat Pinel S, Barberi-Heyob M, Cohen-Jonathan E, Merlin JL, Delmas C, Plenat F, Chastagner P. Erythropoietin-induced reduction of hypoxia before and during fractionated irradiation contributes to improvement of radioresponse in human glioma xenografts. Int J Radiat Oncol Biol Phys. 2004;59(1):250–9.CrossRefPubMed Pinel S, Barberi-Heyob M, Cohen-Jonathan E, Merlin JL, Delmas C, Plenat F, Chastagner P. Erythropoietin-induced reduction of hypoxia before and during fractionated irradiation contributes to improvement of radioresponse in human glioma xenografts. Int J Radiat Oncol Biol Phys. 2004;59(1):250–9.CrossRefPubMed
15.
Zurück zum Zitat Toussaint M, Pinel S, Auger F, Durieux N, Thomassin M, Thomas E, Moussaron A, Meng D, Plénat F, Amouroux M, Bastogne T, Frochot C, Tillement O, Lux F, Barberi-Heyob M. Proton MR spectroscopy and diffusion MR imaging monitoring to predict tumor response to interstitial photodynamic therapy for glioblastoma. Theranostics. 2017;7(2):436–51.CrossRefPubMedPubMedCentral Toussaint M, Pinel S, Auger F, Durieux N, Thomassin M, Thomas E, Moussaron A, Meng D, Plénat F, Amouroux M, Bastogne T, Frochot C, Tillement O, Lux F, Barberi-Heyob M. Proton MR spectroscopy and diffusion MR imaging monitoring to predict tumor response to interstitial photodynamic therapy for glioblastoma. Theranostics. 2017;7(2):436–51.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Hood JD, Cheresh DA. Role of integrins in cell invasion and migration. Nat Rev Cancer. 2002;2(2):91–100.CrossRefPubMed Hood JD, Cheresh DA. Role of integrins in cell invasion and migration. Nat Rev Cancer. 2002;2(2):91–100.CrossRefPubMed
17.
Zurück zum Zitat Nicholson B, Theodorescu D. Angiogenesis and prostate cancer tumor growth. J Cell Biochem. 2004;91(1):125–50.CrossRefPubMed Nicholson B, Theodorescu D. Angiogenesis and prostate cancer tumor growth. J Cell Biochem. 2004;91(1):125–50.CrossRefPubMed
18.
Zurück zum Zitat Al-Husein B, Goc A, Somanath PR. Suppression of interactions between prostate tumor cell-surface integrin and endothelial ICAM-1 by simvastatin inhibits micrometastasis. J Cell Physiol. 2013;228(11):2139–48.CrossRefPubMedPubMedCentral Al-Husein B, Goc A, Somanath PR. Suppression of interactions between prostate tumor cell-surface integrin and endothelial ICAM-1 by simvastatin inhibits micrometastasis. J Cell Physiol. 2013;228(11):2139–48.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Zheng DQ, Woodard AS, Fornaro M, Tallini G, Languino LR. Prostatic carcinoma cell migration via alpha(v)beta3 integrin is modulated by a focal adhesion kinase pathway. Cancer Res. 1999;59(7):1655–64.PubMed Zheng DQ, Woodard AS, Fornaro M, Tallini G, Languino LR. Prostatic carcinoma cell migration via alpha(v)beta3 integrin is modulated by a focal adhesion kinase pathway. Cancer Res. 1999;59(7):1655–64.PubMed
20.
Zurück zum Zitat Maschauer S, Haubner R, Kuwert T, Prante O. (18)F-glyco-RGD peptides for PET imaging of integrin expression: efficient radiosynthesis by click chemistry and modulation of biodistribution by glycosylation. Mol Pharm. 2014;11(2):505–15.CrossRefPubMed Maschauer S, Haubner R, Kuwert T, Prante O. (18)F-glyco-RGD peptides for PET imaging of integrin expression: efficient radiosynthesis by click chemistry and modulation of biodistribution by glycosylation. Mol Pharm. 2014;11(2):505–15.CrossRefPubMed
21.
Zurück zum Zitat Huang Y, Liu W, Gao F, Fang X, Chen Y. c(RGDyK)-decorated Pluronic micelles for enhanced doxorubicin and paclitaxel delivery to brain glioma. Int J Nanomedicine. 2016;11:1629–41.PubMedPubMedCentral Huang Y, Liu W, Gao F, Fang X, Chen Y. c(RGDyK)-decorated Pluronic micelles for enhanced doxorubicin and paclitaxel delivery to brain glioma. Int J Nanomedicine. 2016;11:1629–41.PubMedPubMedCentral
22.
Zurück zum Zitat Yan H, Wang L, Wang J, Weng X, Lei H, Wang X, Jiang L, Zhu J, Lu W, Wei X, Li C. Two-order targeted brain tumor imaging by using an optical/paramagnetic nanoprobe across the blood brain barrier. ACS Nano. 2012;6(1):410–20.CrossRefPubMed Yan H, Wang L, Wang J, Weng X, Lei H, Wang X, Jiang L, Zhu J, Lu W, Wei X, Li C. Two-order targeted brain tumor imaging by using an optical/paramagnetic nanoprobe across the blood brain barrier. ACS Nano. 2012;6(1):410–20.CrossRefPubMed
23.
Zurück zum Zitat Chinnaiyan P, Kensicki E, Bloom G, Prabhu A, Sarcar B, Kahali S, Eschrich S, Qu X, Forsyth P, Gillies R. The metabolomic signature of malignant glioma reflects accelerated anabolic metabolism. Cancer Res. 2012;72(22):5878–88.CrossRefPubMedPubMedCentral Chinnaiyan P, Kensicki E, Bloom G, Prabhu A, Sarcar B, Kahali S, Eschrich S, Qu X, Forsyth P, Gillies R. The metabolomic signature of malignant glioma reflects accelerated anabolic metabolism. Cancer Res. 2012;72(22):5878–88.CrossRefPubMedPubMedCentral
24.
25.
Zurück zum Zitat Higuchi T, Bengel FM, Seidl S, Watzlowik P, Kessler H, Hegenloh R, Reder S, Nekolla SG, Wester HJ, Schwaiger M. Assessment of alphavbeta3 integrin expression after myocardial infarction by positron emission tomography. Cardiovasc Res. 2008;78(2):395–403.CrossRefPubMed Higuchi T, Bengel FM, Seidl S, Watzlowik P, Kessler H, Hegenloh R, Reder S, Nekolla SG, Wester HJ, Schwaiger M. Assessment of alphavbeta3 integrin expression after myocardial infarction by positron emission tomography. Cardiovasc Res. 2008;78(2):395–403.CrossRefPubMed
26.
Zurück zum Zitat Verjans J, Wolters S, Laufer W, Schellings M, Lax M, Lovhaug D, et al. Early molecular imaging of interstitial changes in patients after myocardial infarction: comparison with delayed contrast-enhanced magnetic resonance imaging. J Nucl Cardiol Off Publ Am Soc Nucl Cardiol. 2010;17(6):1065–72.CrossRef Verjans J, Wolters S, Laufer W, Schellings M, Lax M, Lovhaug D, et al. Early molecular imaging of interstitial changes in patients after myocardial infarction: comparison with delayed contrast-enhanced magnetic resonance imaging. J Nucl Cardiol Off Publ Am Soc Nucl Cardiol. 2010;17(6):1065–72.CrossRef
27.
Zurück zum Zitat Beer AJ, Pelisek J, Heider P, Saraste A, Reeps C, Metz S, Seidl S, Kessler H, Wester HJ, Eckstein HH, Schwaiger M. PET/CT imaging of integrin αvβ3 expression in human carotid atherosclerosis. JACC Cardiovasc Imaging. 2014;7(2):178–87.CrossRefPubMed Beer AJ, Pelisek J, Heider P, Saraste A, Reeps C, Metz S, Seidl S, Kessler H, Wester HJ, Eckstein HH, Schwaiger M. PET/CT imaging of integrin αvβ3 expression in human carotid atherosclerosis. JACC Cardiovasc Imaging. 2014;7(2):178–87.CrossRefPubMed
28.
Zurück zum Zitat Choi H, Phi JH, Paeng JC, Kim SK, Lee YS, Jeong JM, Chung JK, Lee DS, Wang KC. Imaging of integrin α(V)β(3) expression using (68)Ga-RGD positron emission tomography in pediatric cerebral infarct. Mol Imaging. 2013;12(4):213–7.CrossRefPubMed Choi H, Phi JH, Paeng JC, Kim SK, Lee YS, Jeong JM, Chung JK, Lee DS, Wang KC. Imaging of integrin α(V)β(3) expression using (68)Ga-RGD positron emission tomography in pediatric cerebral infarct. Mol Imaging. 2013;12(4):213–7.CrossRefPubMed
29.
Zurück zum Zitat Beer AJ, Lorenzen S, Metz S, Herrmann K, Watzlowik P, Wester HJ, Peschel C, Lordick F, Schwaiger M. Comparison of integrin alphaVbeta3 expression and glucose metabolism in primary and metastatic lesions in cancer patients: a PET study using 18F-galacto-RGD and 18F-FDG. J Nucl Med. 2008;49(1):22–9.CrossRefPubMed Beer AJ, Lorenzen S, Metz S, Herrmann K, Watzlowik P, Wester HJ, Peschel C, Lordick F, Schwaiger M. Comparison of integrin alphaVbeta3 expression and glucose metabolism in primary and metastatic lesions in cancer patients: a PET study using 18F-galacto-RGD and 18F-FDG. J Nucl Med. 2008;49(1):22–9.CrossRefPubMed
31.
Zurück zum Zitat Schnell O, Krebs B, Carlsen J, Miederer I, Goetz C, Goldbrunner RH, Wester HJ, Haubner R, Pöpperl G, Holtmannspötter M, Kretzschmar HA, Kessler H, Tonn JC, Schwaiger M, Beer AJ. Imaging of integrin alpha(v)beta(3) expression in patients with malignant glioma by [18F] Galacto-RGD positron emission tomography. Neuro Oncol. 2009;11(6):861–70.CrossRefPubMedPubMedCentral Schnell O, Krebs B, Carlsen J, Miederer I, Goetz C, Goldbrunner RH, Wester HJ, Haubner R, Pöpperl G, Holtmannspötter M, Kretzschmar HA, Kessler H, Tonn JC, Schwaiger M, Beer AJ. Imaging of integrin alpha(v)beta(3) expression in patients with malignant glioma by [18F] Galacto-RGD positron emission tomography. Neuro Oncol. 2009;11(6):861–70.CrossRefPubMedPubMedCentral
Metadaten
Titel
PET imaging of 68Ga-NODAGA-RGD, as compared with 18F-fluorodeoxyglucose, in experimental rodent models of engrafted glioblastoma
verfasst von
Sibel Isal
Julien Pierson
Laetitia Imbert
Alexandra Clement
Charlotte Collet
Sophie Pinel
Nicolas Veran
Aurélie Reinhard
Sylvain Poussier
Guillaume Gauchotte
Steeven Frezier
Gilles Karcher
Pierre-Yves Marie
Fatiha Maskali
Publikationsdatum
01.12.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
EJNMMI Research / Ausgabe 1/2018
Elektronische ISSN: 2191-219X
DOI
https://doi.org/10.1186/s13550-018-0405-5

Weitere Artikel der Ausgabe 1/2018

EJNMMI Research 1/2018 Zur Ausgabe