ArticlePDF Available

Recent Developments of 18F-FET PET in Neuro-oncology

Authors:

Abstract and Figures

Background: From the past decade to date, several studies related to O-(2- [18F]fluoroethyl)-L-tyrosine (18F-FET) positron emission tomography (PET) in brain tumours have been published in the literature. Objective: The aim of this narrative review is to summarize the recent developments and the current role of 18F-FET PET in brain tumours according to recent literature data. Methods: Main findings from selected recently published and relevant articles on the role of 18F-FET PET in neuro-oncology are described. Results: 18F-FET PET may be useful in the differential diagnosis between brain tumours and non-neoplastic lesions and between low-grade and high-grade gliomas. Integration of 18F-FET PET into surgical planning allows better delineation of the extent of resection beyond margins visible with standard MRI. For biopsy planning, 18F-FET PET is particularly useful in identifying malignant foci within non-contrast-enhancing gliomas. 18F-FET PET may improve the radiation therapy planning in patients with gliomas. This metabolic imaging method may be useful to evaluate treatment response in patients with gliomas and it improves the differential diagnosis between brain tumours recurrence and posttreatment changes. 18F-FET PET may provide useful prognostic information in high-grade gliomas. Conclusion: Based on recent literature data 18F-FET PET may provide additional diagnostic information compared to standard MRI in neuro-oncology.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.ae
Current Medicinal Chemistry, 2018, 25, 1-13 1
REVIEW ARTICLE
0929-8673/18 $58.00+.00 © 2018 Bentham Science Publishers
Recent Developments of 18F-FET PET in Neuro-oncology
Barbara Muoio, Luca Giovanella and Giorgio Treglia*
Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
A R T I C L E H I S T O R Y
Received: November 14, 2016
Revised: September 19, 2017
Accepted: November 07, 2017
DOI:
10.2174/0929867325666171123202644
Abstract: Background: From the past decade to date, several studies related to O-(2-
[18F]fluoroethyl)-L-tyrosine (18F-FET) positron emission tomography (PET) in brain tu-
mours have been published in the literature.
Objective: The aim of this narrative review is to summarize the recent developments and
the current role of 18F-FET PET in brain tumours according to recent literature data.
Methods: Main findings from selected recently published and relevant articles on the role
of 18F-FET PET in neuro-oncology are described.
Results: 18F-FET PET may be useful in the differential diagnosis between brain tumours
and non-neoplastic lesions and between low-grade and high-grade gliomas. Integration of
18F-FET PET into surgical planning allows better delineation of the extent of resection
beyond margins visible with standard MRI. For biopsy planning, 18F-FET PET is particu-
larly useful in identifying malignant foci within non-contrast-enhancing gliomas. 18F-FET
PET may improve the radiation therapy planning in patients with gliomas. This metabolic
imaging method may be useful to evaluate treatment response in patients with gliomas
and it improves the differential diagnosis between brain tumours recurrence and post-
treatment changes. 18F-FET PET may provide useful prognostic information in high-grade
gliomas.
Conclusion: Based on recent literature data 18F-FET PET may provide additional diag-
nostic information compared to standard MRI in neuro-oncology.
Keywords: PET, O-(2-[18F]fluoroethyl)-L-tyrosine, FET, neuro-oncology, neuroimaging, glioma, brain tumours,
glioblastoma.
1. INTRODUCTION
Magnetic resonance imaging (MRI) is the standard
neuroimaging method to diagnose neoplastic brain le-
sions, as well as to perform stereotactic biopsy and sur-
gical planning in neuro-oncology. MRI has the advan-
tage of providing structural anatomical details with
high sensitivity, though histological specificity is lim-
ited. Metabolic imaging by using radiolabelled amino
acids may be helpful in the diagnostic evaluation of
brain lesions providing significant information com-
pared to conventional morphological imaging tech-
niques such as computed tomography (CT) or MRI [1].
*Address correspondence to this author at the Department of Nu-
clear Medicine and PET/CT Centre, Oncology Institute of Southern
Switzerland, Via Ospedale 12, zip code: 6500, Bellinzona,
Switzerland; Tel: 0041 918118919; Fax: 0041918118250;
E-mail: giorgio.treglia@eoc.ch
Several factors influence the increased amino acid up-
take in brain tumour cells including the overexpression
of the amino acid transport systems in tumour cells,
alterations in the tumour vasculature and tumour cell
proliferation [1].
O-(2-[18F]fluoroethyl)-L-tyrosine (18F-FET) is a ra-
diolabelled amino acid, tyrosine analogue, which can
be used to visualize brain tumours by using positron
emission tomography (PET). The results of the first
human study using 18F-FET PET in a patient with brain
tumour have been reported in 1999 [2]. Characteristics
of this amino acid radiopharmaceutical are the high in
vivo stability, the fast brain and tumour uptake kinetics,
the usually low accumulation in non-tumour tissue and
its ease of synthesis [2]. 18F-FET penetrates the blood-
brain barrier by a specific amino acid transport system
and it is not incorporated into proteins [2]. Its uptake
2 Current Medicinal Chemistry, 2018, Vol. 25, No. 00 Muoio et al.
mechanism in tumour cells is mediated by the L-type
amino acid transport system [3].
Contraindications for 18F-FET PET are only preg-
nancy and lack of patient cooperation [4].
About the 18F-FET PET imaging procedure [4] pa-
tients should be fasting for more than four hours before
the radiopharmaceutical injection. A mean dose of 200-
250 MBq is intravenously injected in adult patients.
Hybrid PET/CT or PET/MRI is used for image acquisi-
tion. In some centers a 40 minutes dynamic acquisition
just after the radiopharmaceutical injection is per-
formed. In other centers static PET image acquisition
from 20 to 40 minutes post-injection is carried out [4].
18F-FET PET image interpretation is usually performed
by using visual (qualitative) and semi-quantitative cri-
teria such as lesion-to-background uptake ratios
(LBRs) [4].
About dosimetry of 18F-FET PET, the highest ab-
sorbed dose was found for the bladder. The effective
dose is 16.5 microSv/MBq for adults, which would
lead to an effective dose of 6.1 mSv in a PET study
using 370 MBq [5]. The potential radiation risks asso-
ciated with this metabolic imaging method are well
within accepted limits [6].
From the past decade to date, several studies related
to 18F-FET PET in brain tumours have been published
in the literature. The aim of this narrative review is to
summarize the recent developments and the current
role of 18F-FET PET in neuro-oncology according to
selected recently published and relevant articles on this
topic.
2. 18
F-FET PET IN THE INITIAL ASSESSMENT
OF PATIENTS WITH NEWLY DIAGNOSED
BRAIN LESIONS
Recent literature data show the additional high di-
agnostic value of 18F-FET PET in patients with newly
diagnosed brain lesions at MRI [7-24].
2.1. Static Measures
A meta-analysis published in 2012 including 13
studies (462 patients with brain lesions) demonstrated
that 18F-FET PET is an accurate method for the diagno-
sis of primary brain tumour, showing a pooled sensitiv-
ity of 82% and a pooled specificity of 76% [7]. Re-
ceiver-operating-characteristic (ROC) analysis indi-
cated that a mean LBR threshold of at least 1.6 and a
maximum LBR of at least 2.1 had the best diagnostic
value for differentiating primary brain tumours from
non-neoplastic lesions [7]. Furthermore, mean LBR
and maximum LBR were significantly higher in high-
grade gliomas than in low-grade gliomas (mean LBR:
1.7 ± 0.7 vs. 2.6 ± 1.0, respectively; p < 0.001; maxi-
mum LBR: 2.2 ± 0.9 vs. 3.1 ± 1.1, respectively; p <
0.001) [7].
In a recent retrospective study on 174 patients with
newly diagnosed brain lesions on MRI, 18F-FET PET
provided valuable additional information for the differ-
entiation of cerebral lesions and the grading of gliomas.
Neoplastic lesions showed significantly higher 18F-FET
uptake than non-neoplastic lesions (maximum LBR:
3.0 ± 1.3 vs. 1.8 ± 0.5, respectively; p < 0.001). ROC
analysis yielded an optimal cut-off of 2.5 for maximal
LBR to differentiate between neoplastic lesions and
non-neoplastic lesions (sensitivity 57%, specificity
92%, accuracy 62%, positive predictive value 98%, and
negative predictive value 27%). High-grade gliomas
showed significantly higher 18F-FET uptake than low-
grade gliomas (maximal LBR: 3.6 ± 1.4 vs. 2.4 ± 1.0,
respectively; p < 0.001). ROC analysis for differentia-
tion between high-grade and low-grade gliomas yielded
an optimal cut-off of 2.5 for maximum LBR (sensitiv-
ity 80%, specificity 65%, accuracy 72%, positive pre-
dictive value 66%, and negative predictive value 79%).
The results for mean LBR were similar with a cut-off
of 1.9 [10].
Another recent retrospective study on 393 patients
with brain lesions at MRI demonstrated that 18F-FET
PET has a high sensitivity for the detection of high-
grade brain tumours. Its specificity, however, is limited
by passive tracer influx through a disrupted blood-brain
barrier. Sensitivity and specificity of 18F-FET PET for
the detection of brain tumour were 87% and 68%, re-
spectively. Significant differences in maximum LBR
were detected between high-grade brain tumours and
low-grade brain tumours (maximum LBR: 2.04 ± 0.72
and 1.52 ± 0.70, respectively; p < 0.001), as well as
among inflammatory and other non-neoplastic brain
lesions (maximum LBR: 1.66 ± 0.33 and 1.10 ± 0.37,
respectively; p < 0.0001). Gliomas showed 8F-FET up-
take in 80% of World Health Organization (WHO)
grade I, 79% of grade II, 92% of grade III, and 100%
of grade IV tumours. Low-grade oligodendrogliomas
WHO grade II had significantly higher tracer uptakes
than astrocytomas grades II and III (p = 0.018 and p =
0.015, respectively). Furthermore 18F-FET uptake
showed a strong association with contrast enhancement
on MRI (p < 0.001) and was also positive in 52% of
nonglial brain tumours and non-neoplastic brain le-
sions. Nevertheless gliomas showed radiopharmaceuti-
cal uptake in the absence of contrast enhancement on
Recent Developments of 18F-FET PET in Neuro-oncology Current Medicinal Chemistry, 2018, Vol. 25, No. 00 3
MRI, which most likely reflects biologically active tu-
mour [11].
2.2. Kinetic Analysis and Correlation between 18F-
FET PET and MRI-based Parameters
Jansen et al. investigated the discriminative value of
additional dynamic 18F-FET PET in 127 patients with
MRI-suspected low-grade gliomas. Maximum and
mean LBR did not differ significantly between high-
grade and low-grade gliomas but kinetic analysis relia-
bly identified high-grade gliomas (sensitivity 95% and
specificity 72%) [8]. The same group of researchers
demonstrated that 18F-FET uptake was significantly
higher in gliomas with oligodendrial components,
compared with astrocytomas [9].
Zhang et al. explored the relationship between re-
gional cerebral blood flow (rCBF) as measured by arte-
rial spin labelling MRI and the kinetic behaviour of
18F-FET PET in 20 patients with gliomas demonstrat-
ing that there is a relationship between rCBF and 18F-
FET uptake in gliomas in the initial uptake phase, but
the kinetic behaviour of 18F-FET uptake in the late
phase is not significantly influenced by rCBF. Thus,
the differential kinetic pattern of 18F-FET uptake in
high-grade and low-grade gliomas appears to be deter-
mined by factors other than rCBF [12].
The research of Fills et al. by using PET/MRI con-
firmed that in patients with gliomas 18F-FET PET and
regional cerebral blood volume (rCBV) assessed by
perfusion-weighted MR imaging (PWI) yielded differ-
ent information. 18F-FET PET showed considerably
higher maximum LBR and larger tumour volumes than
rCBV maps. The spatial congruence of both parameters
was poor. The locations of the local hot spots differed
considerably. Therefore metabolic active brain tumour
tissue as depicted by 18F-FET PET is not reflected by
rCBV as measured with PWI [13].
The findings from the recent study of Göttler et al.
on 30 untreated glioma patients evaluated with simul-
taneous PET/MRI indicated that 18F-FET PET and
MRI-based rCBV may provide both congruent and
complimentary information on glioma biology [14].
Furthermore as recently published there is no con-
gruency between 18F-FET uptake and diffusivity at dif-
fusion-weighted MRI in nonenhancing low-grade
gliomas. Diffusion restriction in these tumours most
likely represents changes in brain and tumour cell den-
sities as well as alteration of water distribution and is
probably not directly correlated with the density of tu-
mour cells [15].
Combination of dynamic 18F-FET PET and diffu-
sion MRI reached good performance for gliomas grad-
ing. In fact, combination of time-activity curves
(TACs) at dynamic 8F-FET PET and apparent diffusion
coefficient (ADC) histogram analysis at diffusion MRI
improved the sensitivity from 67% to 86% and the
specificity from 63-67% to 100% about the differential
diagnosis between high-grade and low-grade gliomas
[16].
2.3. Dual-time-point PET Imaging
As dynamic 18F-FET PET could be time consuming
compared to static 18F-FET PET, some researchers
demonstrated that dual-time-point 18F-FET PET imag-
ing could be equivalent to dynamic 18F-FET PET for
grading of gliomas [17,18]. Reduced imaging time in-
creases patient comfort and sedation might be avoided.
Furthermore quicker image interpretation with dual-
time-point PET is possible, as no curve evaluation is
needed [17]. As recently demonstrated, early maximum
LBR at 18F-FET PET (with early images performed at
5-15 minutes after radiopharmaceutical injection)
seems to be more accurate for the differentiation be-
tween low-grade and high-grade gliomas than the stan-
dard static PET scan (performed between 20 and 40
minutes after radiopharmaceutical injection) mainly
caused by the characteristic high 18F-FET uptake of
high-grade gliomas in the initial phase. Therefore,
when dynamic 18F-FET PET cannot be performed,
early maximum LBR assessment can be considered as
an alternative for grading of gliomas [19].
2.4. Textural Analysis
Determination of 18F-FET uptake heterogeneity us-
ing textural features recently proved valuable for the
subgrading of high-grade gliomas. In the study by Pyka
et al. on 113 patients with high-grade gliomas all 18F-
FET-PET textural parameters showed the ability to dif-
ferentiate between WHO grade III and IV tumours (ac-
curacy: 77.5%; p<0.001). Further improvement in
discriminatory power was possible through a combina-
tion of texture and metabolic tumour volume, classify-
ing 85% of tumours correctly [20].
2.5. Evaluation of other Brain Tumours Beyond Su-
pratenturial Gliomas in Adult Patients
Beyond gliomas 18F-FET PET may provide addi-
tional information for noninvasive grading of men-
ingiomas as demonstrated by the preliminary study of
Cornelius et al. The difference in maximum LBR be-
tween low-grade and high-grade meningiomas was sig-
4 Current Medicinal Chemistry, 2018, Vol. 25, No. 00 Muoio et al.
nificant in the late phase of 18F-FET uptake (2.1±0.2
vs. 2.5±0.2, respectively; p=0.003) while there
was no significant difference in the early phase. A
maximal LBR cut-off of 2.3 at 18F-FET PET in the late
phase was accurate in differentiating low-grade from
high-grade meningiomas (sensitivity 83%, specificity
83%). Combination of maximum LBR and TAC pat-
tern at dynamic imaging slightly improved the differen-
tiation of high-grade from low-grade meningiomas (ac-
curacy 92%) [21].
Untreated metastases predominantly show increased
18F-FET-uptake. As recently described by Unterrainer
et al. only a third of metastases <1.0 cm was negative
at 18F-FET-PET, most likely due to scanner resolution
and partial volume effects. In metastases >1.0 cm, 18F-
FET-uptake intensity is highly variable and independ-
ent of tumour size. 18F-FET PET might provide addi-
tional information beyond the tumour extent by reflect-
ing molecular features of a metastasis and might be a
useful tool for future clinical applications, e.g. response
assessment [22].
In paediatric patients with newly diagnosed cerebral
lesions, 18F-FET PET may provide useful information
as recently described by Dunkl et al. The highest accu-
racy to detect neoplastic tissue was obtained when the
maximum LBR was 1.7 or greater (sensitivity 79%,
specificity 71%, accuracy 77%) [23].
Finally preliminary data suggest that 18F-FET PET
may add valuable diagnostic information in brainstem
and spinal cord gliomas, particularly when the diagnos-
tic information derived from MRI is equivocal [24].
3. 18F-FET PET FOR GUIDING BIOPSY OF
BRAIN LESIONS AND RESECTION PLANNING
Several recent studies have demonstrated the value
of 18F-FET PET in guiding biopsy and resection plan-
ning in patients with brain lesions at MRI [25-31].
As showed by Floeth et al. 18F-FET PET was more
sensitive to detect glioma tissue than 5-aminolevulinic
acid (5-ALA) fluorescence and should be considered as
an additional tool in resection planning. In fact there
was significant differences between 18F-FET and 5-
ALA uptake in 30 patients with brain lesions sugges-
tive of diffuse WHO grade II or III gliomas on MRI
owing to a limited sensitivity of 5-ALA to detect tu-
mour tissue especially in low-grade gliomas. In biop-
sies corresponding to high-grade gliomas 18F-FET PET
was positive in 86% of cases, but 5-ALA and Gd-
diethylenetriaminepentaacetic acid enhancement on
MRI (Gd) in only 57% of cases. In biopsies corre-
sponding to low-grade gliomas 18F-FET PET was posi-
tive in 41% of cases, while 5-ALA and Gd were nega-
tive in all but one instance. All tumour areas with 5-
ALA fluorescence were positive on 18F-FET PET [25].
A recent cost-effectiveness study demonstrated that
the combined use of 18 F-FET PET and MRI resulted in
an increase of 18.5% in the likelihood of a correct di-
agnosis and that the use 18F-FET PET is cost-effective
for biopsy targeting in patients with glioma [26].
The use of an automated hotspot detection system
through 18F-FET PET uptake data may also be able to
provide more precise information for stereotactic bi-
opsy trajectories compared to MRI-guided plans. The
use of specially designed computational tools may re-
fine surgical planning by improving biopsy targeting
[27].
A combined PET/MRI multimodal imaging ap-
proach may provide potential benefits in detecting
glioma heterogeneity. In fact, based on recent findings,
multimodal imaging-guided stereotactic biopsy corre-
lated more with histological malignancy indices than
targets that were based solely on the highest 18F-FET
uptake or contrast enhancement on MRI [28].
18F-FET PET may be helpful for target selection and
can be integrated in surgical guidance in paediatric pa-
tients with brain tumours. In fact as reported by Misch
et al. 18F-FET-PET image-guided surgical targeting
yielded histological diagnosis with good diagnostic
accuracy in paediatric patients with brain tumours [29].
Finally, it has recently been demonstrated that in pa-
tients with brain metastases surgical treatment planning
based on MRI or 18F-FET PET only might have a sub-
stantial risk of undertreatment at the tumour margins
[31].
4. 18F-FET PET FOR RADIOTHERAPY PLAN-
NING
Several clinical trials have demonstrated the signifi-
cant differences between 18F-FET PET and standard
MRI concerning the definition of tumour volumes for
radiotherapy planning in glioma patients but it remains
open whether 18F-FET PET-based target definition has
a relevant clinical impact for treatment planning [32-
37].
Comparing for radiotherapy planning of glioblas-
toma MRI-based morphological gross tumour volumes
(GTVs) to biological tumour volumes (BTVs) defined
by the abnormal 18F-FET uptake, Niyazi et al. found
that with 18F-FET PET planning the size and geometri-
cal location of GTVs/BTVs differed in the majority of
patients [32].
Recent Developments of 18F-FET PET in Neuro-oncology Current Medicinal Chemistry, 2018, Vol. 25, No. 00 5
Recently Danish researchers assess the impact of
18F-FET PET on the volumetric target definition for
radiation therapy of high-grade gliomas versus the cur-
rent standard using MRI alone. They found that by in-
cluding 18F-FET PET for GTV the clinical target vol-
ume (CTV) increased moderately for most patients, and
quite substantially for a minority of patients. Patients
with glioblastoma were found to be the primary candi-
dates for 18F-FET PET-guided radiation therapy plan-
ning [33].
In a prospective phase II study, 22 patients with
glioblastoma received post-surgical radiochemother-
apy. The radiotherapy was performed as an MRI and
18F-FET PET-based integrated-boost intensity-
modulated radiotherapy (IMRT). The dose escalation
concept with a total dose of 72 Gy, based on 18F-FET-
PET, did not lead to a survival benefit [34].
Rieken et al. investigated the impact of 18F-FET
PET on target volume definition in low- and high-grade
glioma patients undergoing either first or re-irradiation
with particles. Integrating 18F-FET-uptake into the de-
lineation of GTVs yielded larger volumes. Combined
PET- and MRI-derived planning target volumes
(PTVs) were significantly enlarged in high-grade glio-
mas and in case of primary radiation therapy [35].
18F-FET PET may also be useful for guiding re-
irradiation in patients with recurrent high-grade glio-
mas [36].
5. 18F-FET PET FOR DETECTING RESID-
UAL/RECURRENT BRAIN TUMOURS AFTER
TREATMENT
5.1. Detection of Residual Brain Tumours After
Surgery
Recent studies evaluated the usefulness of 18F-FET
PET in detecting residual brain tumour after surgery
[38-40].
Comparing 5-ALA fluorescence and 18F-FET PET
in detecting residual tumour after surgery, Roessler et
al. showed that residual faint 5-ALA uptake was
documented in large areas at the end of glioblastoma
resection and corresponded to tumour infiltration.
These 5-ALA positive resection plans exceeded the
18F-FET uptake areas in postoperative PET scans.
Thus, intraoperative 5-ALA residual fluorescence
seems to be a more sensitive marker than 18F-FET PET
for residual glioblastoma [38].
Otherwise Kläsner et al. demonstrated that early as-
sessment of the resection status in high-grade gliomas
with 18F-FET-PET seems to be feasible. 18F-FET PET
findings were concordant with intra-operative assess-
ment by using 5-ALA and MRI in detecting or exclud-
ing residual high-grade glioma after surgery in 72%
patients, whereas 18F-FET PET revealed discordant
findings in 28% of patients [39].
In a series of 62 patients with high-grade gliomas
postoperative 18F-FET-PET revealed residual tumour
with higher sensitivity than MRI and showed larger
tumour volumes. Performing PET >72 hours after re-
section did not influence PET results. Based on these
finding 18F-FET PET seems a helpful adjunct in addi-
tion to MRI for postoperative assessment of residual
brain tumour [40].
5.2. Differential Diagnosis Between Glioma Recur-
rence and Post-treatment Changes
Several recent studies have underlined the role of
18F-FET PET in differentiating glioma recurrence and
non-neoplastic post-treatment changes [41-46].
In a retrospective study on 124 glioma patients
compared with the diagnostic accuracy of conventional
MRI to diagnose tumour progression or recurrence
(85%), a higher diagnostic accuracy (93%) was
achieved by 18F-FET PET when a mean LBR 2.0 or
time to peak < 45 minutes was present at dynamic PET
imaging (sensitivity 93%, specificity 100%) [41].
18F-FET PET may facilitate the differential diagno-
sis between early tumour progression and pseudopro-
gression following radiochemotherapy of glioblastoma
as demonstrated by Galldiks et al. In patients with
pseudoprogression 18F-FET uptake was significantly
lower than in patients with tumour progression (maxi-
mum LBR: 1.9±0.4 vs. 2.8±0.5, respectively;
mean LBR: 1.8±0.2 vs. 2.3±0.3, respectively;
both p <0.001). A maximum LBR cut-off value of
2.3 had 96% of accuracy in differentiating pseudopro-
gression from tumour progression [42].
18F-FET PET may also provide valuable informa-
tion in differential diagnosis between late pseudopro-
gression and true tumour progression in patients with
glioblastoma. In the study of Kebir et al. maximum
LBR and mean LBR were significantly higher in pa-
tients with tumour progression than in patients with late
pseudoprogression (maximum LBR: 2.4 ± 0.1 vs. 1.5 ±
0.2, respectively; p = 0.003; mean LBR: 2.1 ± 0.1 vs.
1.5 ± 0.2, respectively; p = 0.012). ROC analysis
yielded an optimal cut-off value of 1.9 for maximum
LBR to differentiate between true progression and late
pseudoprogression (sensitivity 84%, specificity 86%,
accuracy 85%) [43].
6 Current Medicinal Chemistry, 2018, Vol. 25, No. 00 Muoio et al.
Furthermore recently it has been demonstrated that
clustering based on textural 18F-FET PET features may
provide valuable information in assessing pseudopro-
gression in high-grade gliomas [44].
Hybrid simultaneous multiparametric 18F-FET
PET/MRI might play a significant role in the evalua-
tion of patients with suspected glioma recurrence as
described by Jena et al. Individually, maximal LBR,
mean LBR, mean apparent diffusion coefficient (AD-
Cmean), and choline-to-creatine (Cho/Cr) ratios as well
as normalized mean relative cerebral blood volume
(rCBVmean) was significant in differentiating recur-
rence from radiation necrosis, with an accuracy of
93.8% for maximum LBR, 87.5% for mean LBR,
81.3% for ADCmean, 96.9% for Cho/Cr ratio, and
90.6% for normalized rCBVmean. Furthermore, the
combination of 18F-FET PET and MRI parameters pro-
vided very high diagnostic accuracy in this setting [45].
5.3. Differential Diagnosis Between Brain Metasta-
ses Recurrence and Post-treatment Changes
18F-FET-PET is also an accurate method in differen-
tiating recurrent brain metastases from radiation necro-
sis as recently demonstrated in several studies [47-50].
In the preliminary study by Galldiks et al. both
maximum LBR and mean LBR were significantly
higher in patients with recurrent metastasis than in pa-
tients with radiation necrosis (maximum LBR: 3.2 ±
0.9 vs. 2.3 ± 0.5, respectively; p < 0.001; mean LBR:
2.1 ± 0.4 vs. 1.8 ± 0.2, respectively; p < 0.001). The
diagnostic accuracy of 18F-FET PET for the correct
identification of recurrent brain metastases reached
78% using maximum LBR with a cut-off of 2.55 (sen-
sitivity 79%, specificity 76%) and 83% using mean
LBR cut-off of 1.95 (sensitivity 74%, specificity 90%).
Diagnostic accuracy increased to 92% by using dy-
namic 18F-FET PET parameters [47].
In a second study including 62 patients LBRs were
significantly higher in recurrent metastases than in ra-
diation injuries (maximum LBR: 3.3 ± 1.0 vs. 2.2 ±
0.4, respectively; p < 0.001; mean LBR: 2.2 ± 0.4 vs.
1.7 ± 0.3, respectively; p < 0.001). The highest accu-
racy (88%) for diagnosing recurrent metastasis could
be obtained with LBRs in combination with dynamic
PET parameters [48].
Another recent study on 50 patients confirmed that
18F-FET uptake was higher in recurrent metastases
compared to radiation-induced changes (maximum
LBR: 2.9 vs. 2.0, respectively; p<0.001; mean LBR:
2.2 vs. 1.7, respectively; p<0.001). Optimal cut-off
values of 2.15 for maximum LBR and 1.95 for mean
LBR gave good diagnostic accuracy (sensitivity 86%,
specificity 79%). Combining LBRs and dynamic PET
parameters, sensitivity and specificity increased to 93%
and 84%, respectively [49].
Lastly, textural feature analysis at 18F-FET PET in
combination with LBRs may have the potential to in-
crease diagnostic accuracy for discrimination between
brain metastases recurrence and radiation injury, with-
out the need for dynamic 18F-FET PET scans, as re-
cently demonstrated by Lohmann et al. [50].
6. 18F-FET PET FOR DETECTING TUMOUR
PROGRESSION AND FOR TREATMENT
MONITORING AND RESPONSE ASSESSMENT
18F-FET PET using both LBRs and kinetic parame-
ters at dynamic imaging may provide valuable diagnos-
tic information for the non-invasive detection of malig-
nant progression of low-grade gliomas with higher di-
agnostic accuracy than changes of contrast enhance-
ment at MRI. Thus, repeated 18F-FET PET may be
helpful for further treatment decisions [51,52].
Treatment monitoring and response assessment in
patients with glioblastoma is difficult by using MRI
because unspecific alterations with contrast enhance-
ment can mimic tumour progression [53].
As demonstrated by the prospective study of
Galldiks et al., in contrast to gadolinium contrast-
enhancement volumes on MRI, changes in 18F-FET
uptake may be a valuable parameter to assess treatment
response in glioblastoma [54].
In recurrent high-grade glioma patients undergoing
antiangiogenic treatment, 18F-FET PET seems to be
useful to predict treatment failure and to provide im-
portant information to response assessment based
solely on MRI [55].
18F-FET PET may also be useful for therapy moni-
toring of glioma after stereotactic iodine-125
brachytherapy; in this setting post-treatment changes
can mimic tumor progression at MRI, whereas 18F-FET
PET performed 6 months after stereotactic brachyther-
apy may differentiate accurately between therapeutic
effects and local tumour progression [56].
18F-FET PET parameters seem to predict bevacizu-
mab-irinotecan treatment failure in patients with recur-
rent high-grade gliomas providing additional informa-
tion for clinical management over and above the infor-
mation obtained by MRI response assessment [57]. The
additional use of 18F-FET PET in the management of
patients with recurrent high-grade gliomas treated with
bevacizumab-irinotecan may be cost-effective. Integra-
Recent Developments of 18F-FET PET in Neuro-oncology Current Medicinal Chemistry, 2018, Vol. 25, No. 00 7
tion of 18F-FET PET has the potential to avoid over-
treatment and corresponding costs, as well as unneces-
sary side effects to the patient [58].
7. PROGNOSTIC ROLE OF 18F-FET PET IN
BRAIN TUMOURS
18F-FET PET can predict prognosis and survival in
patients with gliomas and serves as a valuable tool to
supplement the established clinical and histopathologi-
cal parameters [59].
Absence of 18F-FET uptake in newly diagnosed as-
trocytic low-grade gliomas does not generally indicate
an indolent disease course. Among the 18F-FET-
positive gliomas, decreasing TACs at dynamic 18F-FET
PET may constitute an unfavourable prognostic factor
in astrocytic low-grade gliomas [60]. Another recent
study demonstrated that dynamic 18F-FET PET might
be an important and independent prognostic marker for
patients with grade II glioma [61], whereas no correla-
tion of survival to 18F-FET uptake was observed in
low-grade gliomas in a recent study by Bette et al. [62].
Early time-to-peak at dynamic 18F-FET PET was as-
sociated with worse outcome in patients with newly
diagnosed astrocytic high-grade gliomas [63].
Static 18F-FET PET provided significant and addi-
tional prognostic information in grade III gliomas,
compared to MRI, supporting the use of both modali-
ties preoperatively to assess individual risks and esti-
mate prognosis [64].
A correlation of 18F-FET PET texture but not LBR
was shown with survival of high-grade glioma patients
[20].
Postoperative tumour volume in 18F-FET PET had a
significant independent influence on survival in pa-
tients with glioblastoma [65]. A recent prospective
study demonstrated that 18F-FET PET-derived volumes
before radiochemotherapy and TACs at dynamic 18F-
FET PET represent important prognostic markers in
glioblastoma. 18F-FET PET-derived volume before ra-
diochemotherapy is a strong prognostic factor for sur-
vival independent of the mode of surgery [66].
18F-FET uptake was described as an independent
prognostic determinant in patients with glioma referred
for radiation therapy. Higher 18F-FET uptake appears to
be associated with a worse tumour-related mortality
and a shorter duration of the disease-free interval [67].
Large BTV on 18F-FET PET was shown as independent
prognostic factor of survival in glioblastoma patients
[68].
18F-FET PET was described as a sensitive tool to
predict treatment response in patients with glioblas-
toma at an early stage after radiochemiotherapy. Early
PET responders had a significantly longer median sur-
vival [69]. In contrast to gadolinium contrast enhance-
ment volumes on MRI, changes in 18F-FET PET may
be a valuable parameter to predict survival time [54].
Furthermore 18F-FET PET uptake before re-
irradiation was revealed as independent significant pre-
dictor for survival after re-irradiation of high-grade
gliomas [70-72].
8. COMPARISON OF 18F-FET WITH OTHER
PET RADIOPHARMACEUTICALS FOR BRAIN
TUMOUR IMAGING
Several PET radiopharmaceuticals may be used in
neuro-oncology and some of them have recently been
compared with 18F-FET.
PET using 11C-methionine, a radiolabelled amino
acid tracer, has high sensitivity and specificity for im-
aging of gliomas and brain metastases. The short half-
life of 11C (20 minutes) limits the use of 11C-
methionine PET to institutions with onsite cyclotron.
18F-FET is labelled with 18F (half-life: 120 minutes)
and could be used much more broadly. Based on litera-
ture data 18F-FET PET and 11C-methionine PET pro-
vided comparable diagnostic information on gliomas
and brain metastases [73].
For brain tumour diagnosis radiolabelled choline
PET was not found superior to 18F-FET PET, in par-
ticular in nongadolinium-enhancing low-grade gliomas
[74].
18F-FET PET has shown higher sensitivity in detec-
tion of gliomas than proliferation imaging with 18F-
fluorothymidine (18F-FLT) PET [75]. In high-grade
gliomas 18F-FET PET but not 18F-FLT PET was able to
detect metabolic active tumour tissue beyond contrast
enhancing tumour on MRI. In contrast to 18F-FET,
blood-brain barrier breakdown seems to be a prerequi-
site for 18F-FLT uptake [76].
In evaluating brain tumours PET using another
amino acid tracer, 18F-fluorodihydroxyphenylalanine
(18F-FDOPA), demonstrated superior contrast ratios for
lesions outside the striatum compared to 18F-FET PET,
but 18F-FDOPA uptake values did not correlate with
grading. Compared to 18F-FDOPA, 18F-FET-PET can
provide additional information on tumour grading and
benefits from lower striatal uptake [77]. Whereas visual
analysis revealed no significant differences in uptake
pattern for 18F-FET and 18F-DOPA in patients with
8 Current Medicinal Chemistry, 2018, Vol. 25, No. 00 Muoio et al.
Fig. (1). Axial MRI (a) and 18F-FET PET (b) images in a 54 y.o. female patient with a suspicious right temporal glioma (ar-
row). 18F-FET PET shows an area of increased radiopharmaceutical uptake corresponding to the anterior portion of the cerebral
lesion and suggesting the presence of a component of high-grade glioma, as confirmed by 18F-FET PET-guided biopsy.
primary or recurrent high-grade gliomas both maxi-
mum LBR and mean LBR were significantly higher for
18F-FET PET. However, regarding tumour delineation,
both tracers performed equally well and seem equally
feasible for imaging of primary and recurrent high-
grade gliomas [78].
A recent meta-analysis demonstrated that for brain
tumour diagnosis, 18F-FET PET performed much better
than PET using the glucose analogue 18F-
fluorodeoxyglucose (18F-FDG) and 18F-FET PET
should be preferred when assessing a new isolated
brain tumour. For glioma grading, however, both trac-
ers showed similar performances [79].
GENERAL REMARKS AND CONCLUSIONS
Recommendations for the clinical use of PET in
neuro-oncology have recently been published from the
Response Assessment in Neuro-Oncology (RANO)
working group and the European Association for
Neuro-Oncology (EANO) [80].
Based on evidence-based data 18F-FET PET is an
accurate method in diagnosing brain tumours with
higher accuracy than 18F-FDG PET in this setting [81].
As stated by RANO-EANO recommendations, 18F-FET
PET provides additional diagnostic information for the
management of patients with glioma compared to stan-
dard MRI. In particular, 18F-FET PET improves the
differential diagnosis among brain tumours and and
non-neoplastic lesions and it is useful for glioma grad-
ing [80] (Fig. 1). Although 18F-FET uptake is usually
higher in high-grade compared to low-grade gliomas, a
significant overlap in uptake values may be observed
using static PET measures (maximum and mean LBR).
Kinetic analysis using dynamic 18F-FET PET signifi-
cantly improves the differential diagnosis between low-
grade and high-grade gliomas [80].
Delineation of tumour borders by using 18F-FET
PET is superior compared to standard MRI. Therefore,
it is suggested to integrate 18F-FET PET data for surgi-
cal planning to allow a better delineation of the extent
of resection. As 18F-FET PET may identify malignant
foci within non-contrast-enhancing gliomas, this
method can be used for biopsy planning of gliomas
[80]. 18F-FET PET may also improve the delineation of
a BTV beyond conventional MRI and radiation therapy
planning using 18F-FET PET appears to be feasible
[80].
18F-FET PET may be useful in evaluating treatment
response in patients with high-grade gliomas because a
decrease in 18F-FET uptake and/or volume is associated
with treatment response. Furthermore, 18F-FET PET
may improve the differential diagnosis between brain
tumours recurrence and post-treatment changes com-
pared to standard MRI (Figs. 2 and 3) [80].
About prognosis, 18F-FET uptake is associated with
outcome in high-grade gliomas both in a pretreatment
setting and following therapy [80].
Recent Developments of 18F-FET PET in Neuro-oncology Current Medicinal Chemistry, 2018, Vol. 25, No. 00 9
Fig. (2). Axial MRI (a) and 18F-FET PET (b) images in a 64 y.o. female patient treated with surgery and radiochemotherapy for
a right temporal-occipital high-grade glioma and with a contrast-enhanced area suspicious for disease relapse or radionecrosis
at MRI (arrow). 18F-FET PET shows increased radiopharmaceutical uptake corresponding to the MRI abnormality, suggesting a
disease relapse as confirmed by histology.
Fig. (3). Axial MRI (a, c) and 18F-FET PET (b, d) images in a 58 male patient with a recurrent left temporal high-grade glioma
post-radiochemotherapy, before (a-c) and after (d-f) antiangiogenic therapy. 18F-FET PET after antiangiogenic therapy shows a
new area of increasing radiopharmaceutical uptake in the left temporal lobe (arrows) compared to the pretratment PET scan,
suggesting disease progression.
10 Current Medicinal Chemistry, 2018, Vol. 25, No. 00 Muoio et al.
Unfortunately, to date, the widespread use of 18F-
FET PET is hampered by the limited availability of this
radiopharmaceutical to some countries.
Future perspectives are represented by the stan-
dardization of PET acquisition protocols and the more
widely diffusion of hybrid PET/MRI imaging tech-
nique [82,83].
CONSENT FOR PUBLICATION
Not applicable.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial
or otherwise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCE
[1] Jager, P.L.; Vaalburg, W.; Pruim, J.; de Vries, E.G.; Lan-
gen, K.J.; Piers, D.A. Radiolabeled amino acids: basic as-
pects and clinical applications in oncology. J Nucl Med,
2001, 42(3): 432-445.
[2] Wester, H.J.; Herz, M.; Weber, W.; Heiss, P.; Senekow-
itsch-Schmidtke, R.; Schwaiger, M.; Stöcklin, G. Synthesis
and radiopharmacology of O-(2-[18F]fluoroethyl)-L-
tyrosine for tumor imaging. J Nucl Med, 1999, 40(1): 205-
212.
[3] Heiss, P.; Mayer, S.; Herz, M.; Wester, H.J.; Schwaiger,
M.; Senekowitsch-Schmidtke, R. Investigation of transport
mechanism and uptake kinetics of O-(2-[18F]fluoroethyl)-
L-tyrosine in vitro and in vivo. J Nucl Med, 1999, 40(8):
1367-1373.
[4] Vander Borght, T.; Asenbaum, S.; Bartenstein, P.; Halldin,
C.; Kapucu, O.; Van Laere, K.; Varrone, A.; Tatsch, K.;
European Association of Nuclear Medicine (EANM).
EANM procedure guidelines for brain tumour imaging us-
ing labelled amino acid analogues. Eur J Nucl Med Mol Im-
aging, 2006; 33(11): 1374-1380.
[5] Pauleit, D.; Floeth, F.; Herzog, H.; Hamacher, K.; Tell-
mann, L.; Müller, H.W.; Coenen, H.H.; Langen, K.J.
Whole-body distribution and dosimetry of O-(2-
[18F]fluoroethyl)-L-tyrosine. Eur J Nucl Med Mol Imaging,
2003, 30(4): 519-524.
[6] Tang, G.; Wang, M.; Tang, X.; Luo, L.; Gan, M. Pharma-
cokinetics and radiation dosimetry estimation of O-(2-
[18F]fluoroethyl)-L-tyrosine as oncologic PET tracer. Appl
Radiat Isot, 2003, 58(2): 219-225.
[7] Dunet, V.; Rossier, C.; Buck, A.; Stupp, R.; Prior, J.O. Per-
formance of 18F-fluoro-ethyl-tyrosin e (18F-FET) PET for
the differential diagnosis of primary brain tumor: a system-
atic review and Metaanalysis. J Nucl Med, 2012, 53(2):
207-214.
[8] Jansen, N.L.; Graute, V.; Armbruster, L.; Suchorska, B.;
Lutz, J.; Eigenbrod, S- Cumming, P.; Bartenstein, P.; Tonn,
J.C., Kreth, F.W.; la Fougère, C. MRI-susp ected low-grade
glioma: is there a need to perform dynamic FET PET? Eur
J Nucl Med Mol Imaging, 2012, 39(6): 1021-1029.
[9] Jansen, N.L.; Schwartz, C.; Graute, V.; Eigenbrod, S.; Lutz,
J.; Egensperger, R.; Pöpperl, G.; Kretzschmar, H.A.; Cum-
ming, P.; Bartenstein, P.; Tonn, J.C.; Kreth, F.W.; la
Fougère, C.; Thon, N. Prediction of oligodendroglial histol-
ogy and LOH 1p/19q using dynamic [(18)F]FET-PET im-
aging in intracranial WHO g rade II and III gliomas. Neuro
Oncol, 2012, 14(12): 1473-1480.
[10] Rapp, M.; Heinzel, A.; Galldiks, N.; Stoffels, G.; Felsberg,
J.; Ewelt, C.; Sabel, M.; Steiger, H.J.; Reifenberger, G.;
Beez, T.; Co enen, H.H.; Floeth, F.W.; Langen, K.J. Diag-
nostic performance of 18F-FET PET in newly diagnosed
cerebral lesions suggestiv e of glioma. J Nucl Med, 2013,
54(2): 229-235.
[11] Hutterer, M.; Nowosielski, M.; Putzer, D.; Jansen, N.L.;
Seiz, M.; Schocke, M.; McCoy, M.; Göbel, G.; la Fougère,
C.; Virgolini, I.J.; Trinka, E.; Jacobs, A.H.; Stockhammer,
G. [18F]-fluoro-ethyl-L-tyrosine PET : a valuable diagnostic
tool in neuro-oncology, but not all that glitters is glioma.
Neuro Oncol, 2013, 15(3): 341-351.
[12] Zhang, K.; Langen, K.J.; Neuner, I.; Stoffels, G.; Filss, C.;
Galldiks, N.; Tellmann, L.; Rota Kops, E.; Coenen, H.H.;
Herzog, H.; Shah, N.J. Relationship of regional cerebral
blood flow and kinetic behaviour of O-(2-(18)F-
fluoroethyl)-L-tyrosine uptake in cerebral gliomas. Nucl
Med Commun. 2014, 35(3): 245-251.
[13] Filss, C.P.; Galldiks, N.; Stoffels, G.; Sabel, M.; Wittsack,
H.J.; Turowski, B.; Antoch, G.; Zhang, K.; Fink, G.R.;
Coenen, H.H.; Shah, N.J.; Herzog, H.; Langen, K.J. Com-
parison of 18F-FET PET and perfusion-weighted MR imag-
ing: a PET/MR imaging hybrid study in patients with brain
tumors. J Nucl Med, 2014, 55(4): 540-545.
[14] Göttler, J.; Lukas, M.; Kluge, A.; Kaczmarz, S.; Gempt, J.;
Ringel, F.; Mustafa, M.; Meyer, B.; Zimmer, C.; Schwaiger,
M.; Förster, S.; Preibisch, C.; Pyka, T. Intra-lesional spatial
correlation of st atic and dynamic FET-PET parameters with
MRI-based cerebral blood volume in patients with untreated
glioma. Eur J Nucl Med Mol Imaging, 2017, 44(3): 392-
397.
[15] Rahm, V.; Boxheimer, L.; Bruehlmeier, M.; Berberat, J.;
Nitzsche, E.U.; Remonda, L.; Roelcke, U. Focal changes in
diffusivity on apparent diffusion coefficient MR imaging
and amino acid uptake on PET do not colocalize in nonen-
hancing low-grade gliomas. J Nucl M ed, 2014, 55(4): 546-
550.
[16] Dunet, V.; Maeder, P.; Nicod-Lalonde, M.; Lhermitte, B.;
Pollo, C.; Bloch, J.; Stupp, R., Meuli, R.; Prior, J.O. Com-
bination of MRI and dynamic FET PET for initial glioma
grading. Nuklearmedizin, 2014, 53(4): 155-161.
[17] Lohmann, P.; Herzog, H.; Rota Kops, E.; Stoffels, G.; Ju-
dov, N.; Filss, C.; Galldiks, N.; Tellmann, L.; Weiss, C.;
Sabel, M., Coenen, H.H.; Shah, N.J.; Langen. K.J. Dual-
time-point O-(2-[(18)F]fluoroethyl)-L-tyrosine PET for
grading of cerebral gliomas. Eur Radiol, 2015; 25(10):
3017-3024.
[18] Malkowski, B.; Harat, M.; Zyromska, A.; Wisniewski, T.;
Harat, A.; Lopatto, R.; Furtak, J. The Sum of Tumour-to-
Brain Ratios Improves the Accuracy of Diagnosing Glio-
mas Using 18F-FET PET. PLoS One, 2015, 10(10):
e0140917.
[19] Albert, N.L.; Winkelmann, I.; Suchorska, B.; Wenter, V.;
Schmid-Tannwald, C.; Mille, E.; Todica, A.; Brendel, M.;
Tonn, J.C.; Bartenstein, P.; la Fougère, C. Early static
(18)F-FET-PET scans have a higher accuracy for glioma
grading than the standard 20-40 min scans. Eur J Nucl Med
Mol Imaging. 2016, 43(6): 1105-1114.
[20] Pyka, T.; Gempt, J.; Hiob, D.; Ringel, F.; Schlegel, J.;
Bette, S.; Wester, H.J.; Meyer, B.; Förster, S. Textural
analysis of pre-therapeutic [18F]-FET-PET and its correla-
tion with tumor grade and patient survival in high-grade
Recent Developments of 18F-FET PET in Neuro-oncology Current Medicinal Chemistry, 2018, Vol. 25, No. 00 11
gliomas. Eur J Nucl Med Mol Imaging, 2016, 43(1): 133-
141.
[21] Cornelius, J.F.; Stoffels, G.; Filß, C.; Galldiks, N.; Slotty,
P.; Kamp, M.; el Khatib, M.; Hänggi, D.; Sabel, M.; Fels-
berg, J.; Steiger, H.J.; Coenen, H.H.; Shah, N.J.; Langen,
K.J. Uptake and tracer kinetics of O-(2-(18)F-fluoroethyl)-
L-tyrosine in meningiomas: preliminary results. Eur J Nucl
Med Mol Imaging, 2015, 42(3): 459-467.
[22] Unterrainer, M.; Galldiks, N.; Suchorska, B.; Kowalew,
L.C.; Wenter, V.; Schmid-Tannwald, C.; Niyazi, M.;
Bartenstein, P.; Langen, K.J.; Albert, N.L. 18F-FET PET
uptake characteristics in patients with newly diagnosed and
untreated brain metastasis. J Nucl M ed, 2016, doi: jnu-
med.116.180075.
[23] Dunkl, V.; Cleff, C.; Stoffels, G.; Judov, N.; Sarikaya-
Seiwert, S.; Law, I.; Bøgeskov, L.; Nysom, K.; Andersen,
S.B.; Steiger, H.J.; Fink, G.R.; Reifenberger, G.; Shah, N.J.;
Coenen, H.H.; Langen, K.J.; Galldiks, N. The usefulness of
dynamic O-(2-18F-fluoroethyl)-L-tyrosine PET in the clini-
cal ev aluation of brain tumors in children and adolescents. J
Nucl Med, 2015, 56(1):88-92.
[24] Tscherpel, C.; Dunkl, V.; Ceccon, G.; Stoffels, G.; Judov,
N.; Rapp, M.; Meyer, P.T.; Kops, E.R.; Ermert, J.; Fink,
G.R.; Shah, N.J.; Langen, K.J.; Galldiks, N. The use of O-
(2-18F-fluoroethyl)-L-tyrosine PET in the diagnosis of
gliomas located in the brainstem and spinal cord. Neuro
Oncol, 2016, doi: 10.1093/neuonc/now243.
[25] Floeth, F.W.; Sabel, M.; Ewelt, C.; Stummer, W.; Felsberg,
J.; R eifenberger, G.; Steiger, H.J.; Sto ffels, G.; Coen en,
H.H.; Langen, K.J. Comparison of (18)F-FET PET and 5-
ALA fluorescence in cerebral gliomas. Eur J Nucl Med Mol
Imaging, 2011, 38(4): 731-741.
[26] Heinzel, A.; Stock, S.; Langen, K.J.; Müller, D. Cost-
effectiveness analysis of FET PET-guided target selection
for th e diagnosis of gliom as. Eur J Nucl Med Mol Imaging,
2012, 39(7): 1089-1096.
[27] Reithmeier, T.; Cordeiro, J.; Mix, M.; Trippel, M.; Rotten-
burger, C.; Nikkhah, G. Impact of automated hotspot detec-
tion for (18)FET PET-guided stereotactic biopsy. Acta Neu-
rochir Suppl, 2013; 117: 93-99.
[28] Gempt, J.; Soehngen, E.; Förster, S.; Ryang, Y.M.;
Schlegel, J.; Zimmer, C.; Meyer, B.; Ringel, F.; Grams,
A.E.; Förschler, A. Multimodal imaging in cerebral gliomas
and its neuropathological correlation. Eur J Radiol, 2014,
83(5): 829-834.
[29] Misch, M.; Guggemos, A.; Driever, P.H.; Koch, A.; Grosse,
F.; Steffen, I.G.; Plotkin, M.; Thomale, U.W. (18)F-FET-
PET guided surgical biopsy and resection in children and
adolescence with brain tumors. Childs Nerv Syst, 2015,
31(2): 261-267.
[30] Lopez, W.O.; Cordeiro, J.G.; Albicker, U.; Doostkam, S.;
Nikkhah, G.; Kirch, R.D. Trippel, M.; Reithmeier, T. Corre-
lation of (18)F-fluoroethyl tyrosine positron-emission to-
mography uptake values and histomorphological findings
by stereotactic serial biopsy in newly diagnosed brain tu-
mors using a refined software tool. Onco Targets Ther,
2015, 17(8): 3803-3815.
[31] Gempt, J.; Bette, S.; Buchmann, N.; Ryang, Y.M.; För-
schler, A.; Pyka, T.; Wester, H.J.; Förster, S.; Meyer, B.;
Ringel, F. Volumetric Analysis of F-18-FET-PET Imaging
for Brain Metastases. World Neurosurg, 2015, 84(6): 1790-
1797.
[32] Niyazi, M.; Geisler, J.; Siefert, A.; Schwarz, S.B.; Gan-
swindt, U.; Garny, S.; Schnell, O.; Suchorska, B.; Kreth,
F.W.; Tonn, J.C.; Bartenstein, P.; la Fougère, C.; Belka, C.
FET-PET for malignant glioma treatment planning. Radio-
ther Oncol, 2011, 99(1): 44-48.
[33] Munck Af Rosenschold, P.; Costa, J.; Engelholm, S.A.;
Lundemann, M.J.; Law, I.; Ohlhues, L.; Engelholm, S. Im-
pact of [18F]-fluoro-ethyl-tyrosine PET imaging on target
definition for radiation therapy of high-grade glioma. Neuro
Oncol, 2015, 17(5): 757-763.
[34] Piroth, M.D.; Pinkawa, M.; Holy, R.; Klotz, J.; Schaar, S.;
Stoffels, G.; Galldiks, N.; Coenen, H.H.; Kaiser, H.J.; Lan-
gen, K.J.; Eble, M.J. Integrated boost IMRT with FET-
PET-adapted local dose escalation in glioblastomas. Results
of a prospective phase II study. Strahlenther Onkol, 2012,
188(4): 334-339.
[35] Rieken, S.; Habermehl, D.; Giesel, F.L.; Hoffmann, C.;
Burger, U.; Rief, H.; Welzel, T.; Haberkorn, U.; Debus, J.;
Combs, S.E. Analysis of FET-PET imaging for target vol-
ume definition in patients with gliomas treated with con-
formal radiotherapy. Radiother Oncol, 2013, 109(3): 487-
492.
[36] Piroth, M.D.; Galldiks, N.; Pinkawa, M.; Holy, R.; Stoffels,
G.; Ermert, J.; Mottaghy, F.M.; Shah, N.J.; Langen, K.J.;
Eble, M.J. Relapse patterns after radiochemotherapy of
glioblastoma with FET PET-guided boost irradiation and
simulation to optimize radiation target volume. Radiat On-
col, 2016, 11: 87.
[37] Lundemann, M.; Costa, J.C.; Law, I.; Engelholm, S.A.;
Muhic, A.; Poulsen, H.S.; Munck Af Rosenschold, P. Pat-
terns of failure for patients with glioblastoma following O-
(2-[(18)F]fluoroethyl)-L-tyrosine PET- and MRI-guided ra-
diotherapy. Radiother Oncol, 2017, 122(3): 380-386.
[38] Roessler, K.; Becherer, A.; Donat, M.; Cejna, M.; Zachen-
hofer, I. Intraoperative tissue fluorescence using 5-
aminolevolinic acid (5-ALA) is more sensitive than contrast
MRI or amino acid positron emission tomography ((18)F-
FET PET) in glioblastoma surgery. Neurol Res, 2012,
34(3): 314-317.
[39] Kläsner, B.; Buchmann, N.; Gempt, J.; Ringel, F.; Lapa, C.;
Krause, B.J. Early [18F]FET-PET in Gliomas after Surgical
Resection: Comparison with MRI and Histopathology.
PLoS One, 2015, 10(10): e0141153.
[40] Buchmann, N.; Kläsner, B.; Gempt, J.; Bauer, J.S.; Pyka,
T.; Delbridge, C.; Meyer, B.; Krause, B.J.; Ringel, F. (18)F-
Fluoroethyl-l-Thyrosine Positron Emission Tomography to
Delineate Tumor Residuals After Glioblastoma Resection:
A Comparison with Standard Postoperative Magnetic
Resonance Imaging. World Neurosurg, 2016, 89: 420-426.
[41] Galldiks, N.; Stoffels, G.; Filss, C.; Rapp, M.; Blau, T.;
Tscherpel, C.; Ceccon, G.; Dunkl, V.; Weinzierl, M.; Stof-
fel, M.; Sab el, M.; Fink, G.R.; Shah, N.J.; Langen, K.J. The
use of dynamic O-(2-18F-fluoroethyl)-l-tyrosine PET in the
diagnosis of patients with progressive and recurrent glioma.
Neuro Oncol, 2015; 17(9): 1293-1300.
[42] Galldiks, N.; Dunkl, V.; Stoffels, G.; Hutterer, M.; Rapp,
M.; Sabel, M.; Reifenberger, G.; Kebir, S.; Dorn, F.; Blau,
T.; Herrlinger, U.; Hau, P.; Ruge, M.I.; Kocher, M.; Gold-
brunner, R.; Fink, G.R.; Drzezga, A.; Schmidt, M.; Langen,
K.J. Diagnosis of pseudoprogression in patients with
glioblastoma using O-(2-[18F]fluoroethyl)-L-tyrosine PET.
Eur J Nucl Med Mol Imaging, 2015, 42(5): 685-695.
[43] Kebir, S.; Fimmers, R.; Galldiks, N.; Schäfer, N.; Mack, F.;
Schaub, C.; Stuplich, M.; Niessen, M.; T zaridis, T.; Simon,
M.; Stoffels, G.; Langen, K.J.; Scheffler, B.; Glas, M.;
Herrlinger, U. Late Pseudoprogression in Glioblastoma: Di-
agnostic Value of Dynamic O-(2-[18F]fluoroethyl)-L-
Tyrosine PET. Clin Cancer Res, 2016, 22(9): 2190-2196.
[44] Kebir, S.; Khurshid, Z.; Gaertner, F.C.; Essler, M.; Hattin-
gen, E.; Fimmers, R.; Scheffler, B.; Herrlinger, U.; Bund-
schuh, R.A.; Glas, M. Unsupervised consensus cluster
analysis of [18F]-fluoroethyl-L-tyrosine positron emission
tomography identified textural features for the diagnosis of
12 Current Medicinal Chemistry, 2018, Vol. 25, No. 00 Muoio et al.
pseudoprogression in high-grade glioma. Oncotarget, 2017,
8(5): 8294-8304.
[45] Jena, A .; Taneja, S.; Gambhir, A. ; Mishra, A.K.; Dʼsouza,
M.M.; Verma, S.M.; H azari, P.P.; Negi, P.; Jhadav, G.K.;
Sogani, S.K. Glioma Recurrence Versus Radiation Necro-
sis: Single-Session Multiparametric Approach Using Simul-
taneous O-(2-18F-Fluoroethyl)-L-Tyrosine PET/MRI. Clin
Nucl Med, 2016, 41(5): e228-236.
[46] Sogani, S.K.; Jena, A.; Taneja, S.; Gambhir, A.; Mishra,
A.K.; D'Souza, M.M.; Verma, S.M.; Hazari, P.P.; Negi, P.;
Jadhav, G.K. Potential for differentiation of glioma recur-
rence from radionecrosis using integrated (18)F-
fluoroethyl-L-tyrosine (FET) positron emission tomogra-
phy/magnetic resonance imaging: A prospective evaluation.
Neurol India, 2017, 65(2): 293-301.
[47] Galldiks, N.; Stoffels, G.; Filss, C.P.; Piroth, M.D.; Sabel,
M.; Ruge, M.I.; Herzog, H.; Shah, N.J.; Fink, G.R.; Coe-
nen, H.H.; Langen, K.J. Role of O-(2-(18)F-fluoroethyl)-L-
tyrosine PET for differentiation of local recurrent brain me-
tastasis from radiation necrosis. J Nucl Med, 2012, 53(9):
1367-1374.
[48] Ceccon, G.; Lohmann, P.; Stoffels, G.; Judov, N.; Filss,
C.P.; Rapp, M.; Bauer, E.; Hamisch, C.; Ruge, M.I.; Ko-
cher, M.; Kuchelmeister, K.; S ellhaus, B.; Sabel, M.; Fink,
G.R.; Shah, N.J.; Langen, K.J.; Galldiks, N. Dynamic O-(2-
18F-fluoroethyl)-L-tyrosine positron emission tomography
differentiates brain metastasis recurrence from radiation in-
jury after radiotherapy. Neuro Oncol, 2016, doi:
10.1093/neuonc/now149.
[49] Romagna, A.; Unterrainer, M.; Schmid-Tannwald, C.;
Brendel, M.; Tonn, J.C.; Nachbichler, S.B.; Muacevic, A.;
Bartenstein, P.; Kreth, F.W.; Albert, N.L. Suspected recur-
rence of b rain metastases after focused high dose radiother-
apy: can [(18)F]FET- PET overcome diagnostic uncertain-
ties? Radiat Oncol, 2016, 11(1): 139.
[50] Lohmann, P.; Stoffels, G.; Ceccon, G.; Rapp, M.; Sabel,
M.; Filss, C.P.; Kamp, M.A.; Stegmayr, C.; Neumaier, B.;
Shah, N.J.; Langen, K.J.; Galldiks, N. Radiation injury vs.
recurrent brain metastasis: combining textural feature ra-
diomics analysis and standard parameters may increase
(18)F-FET PET accuracy without dynamic scans. Eur Ra-
diol, 2016, doi: 10.1007/s00330-016-4638-2.
[51] Galldiks, N.; Stoffels, G.; Ruge, M.I.; Rapp, M.; Sabel, M.;
Reifenberger, G.; Erdem, Z.; Shah, N.J.; Fink, G.R.; Coe-
nen, H.H.; Langen, K.J. Role of O-(2-18F-fluoroethyl)-L-
tyrosine PET as a diagnostic tool for detection of malignant
progression in patients with low-grade glioma. J Nucl Med.
2013, 54(12): 2046-2054.
[52] Unterrainer, M.; Schweisthal, F.; Suchorska, B.; Wenter,
V.; Schmid-Tannwald, C.; Fendler, W.P.; Schüller, U.;
Bartenstein, P.; Tonn, J.C.; Albert, N.L. Serial 18F-FET
PET Imaging of Primarily 18F-FET-Negative Glioma:
Does It Make Sense? J Nucl Med, 2016, 57(8): 1177-1182.
[53] Piroth, M.D.; Liebenstund, S.; Galldiks, N.; Stoffels, G.;
Shah, N.J.; Eble, M.J.; Coenen, H.H.; Langen, K.J. Moni-
toring of radiochemotherapy in patients with glioblastoma
using O-(2-¹Fluoroethyl)-L-tyrosine positron emission
tomography: is dynamic imaging helpful? Mol Imaging,
2013, 12(6): 388-395.
[54] Galldiks, N.; Langen, K.J.; Holy, R.; Pinkawa, M.; Stoffels,
G.; Nolte, K.W.; Kaiser, H.J.; Filss, C.P.; Fink, G.R.; Coe-
nen, H.H.; Eble, M.J.; Piroth, M.D. Assessment of treat-
ment response in patients with glioblastoma using O-(2-
18F-fluoroethyl)-L-tyrosine PET in comparison to MRI. J
Nucl Med, 2012, 53(7): 1048-1057.
[55] Hutterer, M.; Nowosielski, M.; Putzer, D.; Waitz, D.; Tink-
hauser, G.; Kostron, H.; Muigg, A.; Virgolini, I.J.; Staffen,
W.; Trinka, E.; Gotwald, T.; Jacobs, A.H.; Stockhammer,
G. O-(2-18F-fluoroethyl)-L-tyrosine PET predicts failure of
antiangiogeni c treatment in patients with recurrent high-
grade glioma. J Nucl Med, 2011, 52(6):856-864.
[56] Jansen, N.L.; Suchorska, B.; Schwarz, S.B.; Eigenbrod, S.;
Lutz, J.; Graute, V.; Bartenstein, P.; Belka, C.; Kreth, F.W.;
la Fougère, C. [18F]fluoroethyltyrosine-positron emission
tomography-based therapy monitoring after stereotactic io-
dine-125 brachytherapy in patients with recurrent high-
grade glioma. Mol Imaging, 2013, 12(3): 137-147.
[57] Galldiks, N,; Rapp, M.; Stoffels, G.; Fink, G.R.; Shah, N.J.;
Coenen, H.H.; Sabel, M.; Langen, K.J. Response assess-
ment of bevacizumab in patients with recurrent malignant
glioma using [18F]Fluoroethyl-L-tyrosine PET in compari-
son to MRI. Eur J Nucl Med Mol Imaging, 2013, 40(1): 22-
33.
[58] Heinzel, A.; Müller, D.; Langen, K.J.; Blaum, M.; Verburg,
F.A.; Mottaghy, F.M.; Galldiks, N. The use of O-(2-18F-
fluoroethyl)-L-tyrosine PET for treatment management of
bevacizumab and irinotecan in patients with recurrent high-
grade glioma: a cost-effectiveness analysis. J Nucl Med,
2013, 54(8): 1217-1222.
[59] Gempt, J.; Bette, S.; Ryang, Y.M.; Buchmann, N.; Peschke,
P.; Pyka, T.; Wester, H.J.; Förster, S.; Meyer, B.; Ringel, F.
18F-fluoro-ethyl-tyrosine positron emission tomography for
grading and estimation of prognosis in patients with intrac-
ranial gliomas. Eur J Radiol. 2015, 84(5): 955-962.
[60] Jansen, N.L.; Suchorska, B.; Wenter, V.; Eigenbrod, S.;
Schmid-Tannwald, C.; Zwergal, A.; Niyazi, M.; Drexler,
M.; Bartenstein, P.; Schnell, O.; Tonn, J.C.; Thon, N.;
Kreth, F.W.; la Fougère, C. Dynamic 18F-FET PET in
newly diagnosed astrocytic low-grade glioma identifies
high-risk patients. J Nucl Med, 2014, 55(2): 198-203.
[61] Thon, N.; Kunz, M.; Lemke, L.; Jansen, N.L.; Eigenbrod,
S.; Kreth, S.; Lutz, J.; Egensperger, R.; Giese, A.; Herms,
J.; Weller, M.; Kretzschm ar, H .; Tonn, J.C.; la Fougère, C.;
Kreth, F.W. Dynamic 18F-FET PET in suspected WHO
grade II gliomas defines distinct biological subgroups with
different clinical courses. Int J Cancer, 2015, 136(9): 2132-
2145.
[62] Bette, S.; Gempt, J.; Delbridge, C.; Kirschke, J.S.; Schlegel,
J.; Foerster, S.; Huber, T.; Pyka, T.; Zimmer, C .; Meyer, B.;
Ringel, F. Prognostic Value of O-(2-[18F]-Fluoroethyl)-L-
Tyrosine-Positron Emission Tomography Imaging for
Histopathologic Characteristics and Progression-Free Sur-
vival in Patients with Low-Grade Glioma. World Neuro-
surg. 2016, 89: 230-239.
[63] Jansen, N.L.; Suchorska, B.; Wenter, V.; Schmid-
Tannwald, C.; Todica, A.; Eigenbrod, S.; Niyazi, M.; Tonn,
J.C.; Bartenstein, P.; K reth, F.W.; la Fougère, C. Prognostic
significance of dynamic 18F-FET PET in newly diagnosed
astrocytic high-grade glioma. J Nucl Med, 2015, 56(1): 9-
15.
[64] Bette, S.; Peschke, P.; Kaesmacher, J.; Delbridge, C.; Pyka,
T.; Schmidt-Graf, F.; Zimmer, C.; Meyer, B.; Ringel, F.;
Gempt, J. Static FET-PET and MR Imaging in Anaplastic
Gliomas (WHO III). World Neurosurg, 2016, 91: 524-531.
[65] Suchorska, B.; Jansen, N.L.; Linn, J.; Kretzschmar, H.;
Janssen, H.; Eigenbrod, S.; Simon, M.; Pöpperl, G.; Kreth,
F.W.; la Fougere, C.; Weller, M.; Tonn, J.C.; German
Glioma Network. Biological tumor volume in 18FET-PET
before radiochemotherapy correlates with survival in GBM.
Neurology, 2015, 84(7): 710-719.
[66] Piroth, M.D.; Holy, R.; Pinkawa, M.; Stoffels, G.; Kaiser,
H.J.; Galldiks, N.; Herzog, H.; Coenen, H.H.; Eble, M.J.;
Langen, K.J. Prognostic impact of postoperative, pre-
irradiation (18)F-fluoroethyl-l-tyrosine uptake in glioblas-
toma patients treated with radiochemoth erapy. Radiother
Oncol, 2011, 99(2): 218-224.
Recent Developments of 18F-FET PET in Neuro-oncology Current Medicinal Chemistry, 2018, Vol. 25, No. 00 13
[67] Sweeney, R.; Polat, B.; Samnick, S.; Reiners, C.; Flentje,
M.; Verburg, F.A. O-(2-[(18)F]fluoroethyl)-L-tyrosine up-
take is an independent prognostic determinant in patients
with glioma referred for radiation therapy. Ann Nucl Med,
2014, 28(2): 154-162.
[68] Poulsen, S.H.; Urup, T.; Grunnet, K.; Christensen, I.J.; Lar-
sen, V.A.; Jensen, M.L.; Af Rosenschöld, P.M.; Poulsen,
H.S.; Law, I. The prognostic value of FET PET at radio-
therapy planning in newly diagnosed glioblastoma. Eur J
Nucl Med Mol Imaging. 2017, 44(3): 373-381.
[69] Piroth, M.D.; Pinkawa, M.; Holy, R.; Klotz, J.; Nussen, S.;
Stoffels, G.; Coenen, H.H.; Kaiser, H.J.; Langen, K.J.; Eble,
M.J. Prognostic value of early [18F]fluoroethyltyrosine
positron emission tomography after radiochemotherapy in
glioblastoma multiforme. Int J Radiat Oncol Biol Phys,
2011, 80(1): 176-184.
[70] Niyazi, M.; Jansen, N.; Ganswindt, U.; Schwarz, S.B.;
Geisler, J.; Schnell, O.; Büsing, K.; Eigenbrod, S.; la
Fougère, C.; Belka, C. Re-irradiation in recurrent malignant
glioma: prognostic value of [18F]FET-PET. J Neurooncol.
2012, 110(3): 389-395.
[71] Moller, S.; Law, I.; Munck Af Rosenschold, P.; Costa, J.;
Poulsen, H.S.; Engelholm, S.A.; Engelholm, S. Prognostic
value of (18)F-FET PET imaging in re-irradiation of high-
grade glioma: Results of a phase I clinical trial. Radiother
Oncol, 2016, 121(1): 132-137.
[72] Fleischmann, D.F.; Unterrainer, M.; Bartenstein, P.; Belka,
C.; Albert, N.L.; Niyazi, M. (18)F-FET PET prior to recur-
rent high-grade glioma re-irradiation-additional prognostic
value of dynamic time-to-peak analysis and early static
summation images? J Neurooncol, 2017 doi:
10.1007/s11060-016-2366-8.
[73] Grosu, A.L.; Astner, S.T.; Riedel, E.; Nieder, C.; Wieden-
mann, N.; Heinemann, F.; S chwaiger, M; Molls, M.;
Wester, H.J.; Weber, W.A. An interindividual comparison
of O-(2-[18F]fluoroethyl)-L-tyrosine (FET)- and L-[methyl-
11C]methionine (MET)-PET in patients with brain gliomas
and metastases. Int J Radiat Oncol Biol Phys. 2011,
81(4):1049-1058.
[74] Roelcke, U.; Bruehlmeier, M.; Hefti, M.; Hundsberger, T.;
Nitzsche, E.U. F-18 choline PET does not detect increased
metabolism in F-18 fluoroethyltyrosine-negative low-grade
gliomas. Clin Nucl Med. 2012, 37(1): e1-3.
[75] Jeong, S.Y.; Lim, S.M. Comparison of 3'-deoxy-3'-
[18F]fluorothymidine PET and O-(2-[18F]fluoroethyl)-L-
tyrosine PET in patients with newly diagnosed glioma. Nucl
Med Biol, 2012, 39(7): 977-981.
[76] Nowosielski, M.; DiFranco, M.D.; Putzer, D.; Seiz, M.;
Recheis, W.; Jacobs, A.H.; Stockhammer, G.; Hutterer, M.
An intra-individual comparison of MRI, [18F]-FET and
[18F]-FLT PET in patients with high-grade gliomas. PLoS
One, 2014, 9(4): e95830.
[77] Kratochwil, C.; Combs, S.E.; Leotta, K.; Afshar-Oromieh,
A.; Rieken, S.; Debus, J.; Haberkorn, U.; Giesel, F.L. Intra-
individual comparison of ¹F-FET and ¹F-DOPA in PET
imaging of recurrent b rain tumors. Neuro Oncol, 2014,
16(3):434-440.
[78] Lapa, C.; Linsenmann, T.; Monoranu, C.M.; Samnick, S.;
Buck, A.K.; Bluemel, C.; Czernin, J.; Kessler, A.F.; Ho-
mola, G.A.; Ernestus, R.I.; Löhr, M.; Herrmann, K. Com-
parison of the amino acid tracers 18F-FET and 18F-DOPA
in high-grade glioma patients. J Nucl Med, 2014, 55(10):
1611-1616.
[79] Dunet, V.; Pomoni, A.; Hottinger, A.; Nicod-Lalonde, M.;
Prior, J.O. Performance of 18F-FET versus 18F-FDG-PET
for the diagnosis and grading of brain tumors: systematic
review and meta-analysis. Neuro Oncol, 2016, 18(3): 426-
434.
[80] Albert, N.L.; Weller, M.; Suchorska, B.; Galldiks, N.; Sof-
fietti, R.; Kim, M.M.; la Fougère, C.; Pope, W.; Law, I.;
Arbizu, J.; Chamberlain, M.C.; Vogelbaum, M.; Ellingson,
B.M.; Tonn, J.C. Response Assessment in Neuro-Oncology
working group and European Association for Neu ro-
Oncology recommendations for the clinical use of PET im-
aging in gliomas. Neuro Oncol. 2016, 18(9): 1199-1208.
[81] Treglia, G.; Sadeghi, R.; Del Sole, A.; Giovanella, L. Di ag-
nostic performance of PET/CT with tracers other than F-18-
FDG in oncology: an evidence-based review. Clin Transl
Oncol, 2014, 16(9): 770-775.
[82] Henriksen, O.M.; Larsen, V.A.; Muhic, A.; Hansen, A.E.;
Larsson, H.B.; Poulsen, H.S.; Law, I. Simultaneous evalua-
tion of brain tumour metabolism, structure and blood vol-
ume using [(18)F]-fluoroethyltyrosine (FET) PET/MRI:
feasibility, agreement and initial experience. Eur J Nucl
Med Mol Imaging, 2016, 43(1): 103-112.
[83] Ferda, J.; Ferdová, E.; Hes, O.; Mraček, J.; Kreuzberg, B.;
Baxa, J. PET/MRI: Multiparametric imaging of brain tu-
mors. Eur J Radiol, 2017 doi: 10.1016/j.ejrad.2017.02.034.
DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The
Editorial Department reserves the right to make minor modifications for further improvement of the manuscript.
PMID: 29173147
... MRI is the standard neuroimaging method used for diagnosis of brain tumors, for performing stereotactic biopsy, and for surgical planning in neuro-oncology [35]. Current evidence based data also suggest that radiolabeled amino acid PET or PET/CT is an accurate diagnostic method for several clinical indications including evaluation of suspicious brain tumors, glioma grading and delineation, detection of brain tumor recurrence and in providing useful prognostic information in patients with brain tumors [36,37]. ...
Article
Full-text available
Purpose of Review While primary headaches like migraines or cluster headaches are prevalent and often debilitating, it's the secondary headaches—those resulting from underlying pathologies—that can be particularly ominous. This article delves into the sinister causes of headaches, underscoring the importance of a meticulous clinical approach, especially when presented with red flags. Recent Findings Headaches, one of the most common complaints in clinical practice, span a spectrum from benign tension-type episodes to harbingers of life-threatening conditions. For the seasoned physician, differentiating between these extremes is paramount. Summary Headache etiologies covered in this article will include subarachnoid hemorrhage (SAH), cervical artery dissection, cerebral venous thrombosis, meningitis, obstructive hydrocephalus, and brain tumor.
... Neurology continues to be one of the foremost areas in which functional imaging can provide unique information, both for clinical and research purposes. PET/CT or PET/MRI, which provide both functional and morphological information, may be useful for detecting brain tumors in the early stages [34]. While FDG continues to be the most common radiotracer for the evaluation of glucose metabolism, alternative radiotracers have been developed over the last few years [35]. ...
... It penetrates through the blood-brain barrier via a particular amino acid transport pathway and is not incorporated into proteins. This also allows it to be highly stable and rapid uptake by the brain tumors [37]. Another amino acid radiotracer, [ 11 C]MET, which has the same uptake mechanism as [ 18 F]FET, has also been shown in many existing literatures to be more effective than [ [38,40]. ...
Article
Full-text available
PurposeOur systematic review and meta-analysis aimed to evaluate the diagnostic performance of [18F]FDG PET/MRI vs. [18F]FET PET/MRI for recurrent glioma.Methods We searched for relevant articles in PubMed, Embase, and Web of Science until February 2023. Studies regarding the diagnostic performance of [18F]FET PET/MRI or [18F]FDG PET/MRI in recurrent glioma were included. The quality of each study was assessed using the Quality Assessment of Diagnostic Performance Studies-2 (QUADAS-2) tool.ResultsA total of nine studies with 310 patients were included in the analysis, the pooled sensitivity, specificity of [18F]FDG PET/MRI in detecting recurrent glioma after definitive treatment were 0.99 (95% CI 0.75–1.00) and 0.62 (95% CI 0.37–0.83), [18F]FET PET/MRI were 0.94 (95% CI 0.81–0.98) and 0.87 (95% CI 0.70–0.95), respectively. The area under curve (AUC) for [18F]FDG PET/MRI and [18F]FET PET/MRI were 0.92 (95% CI 0.89–0.94) and 0.96 (95% CI 0.94–0.97) and there is a statistical difference between the two imaging modalities (Z = 12.433, P < 0.001). Fagan nomogram indicated that when the pre-test probability was set at 50%, the post-test probability for [18F]FDG PET/MRI and [18F]FET PET/MRI could increase to 72 and 88%.Conclusions[18F]FET PET/MRI has a higher AUC compared to [18F]FDG PET/MRI in the detection of tumor recurrence in glioma. However, the results of the meta-analysis were drawn from studies with small samples. Further larger prospective studies in this setting are warranted.
... In combination with other imaging features, such as calcifications on computer tomography or apparent diffusion coefficient (ADC) or cerebral blood volume (CBV) on MRI, efforts have been made to increase the sensitivity and specificity of non-invasive tests to predict the glial subtype or IDH mutation status but have yielded varying results [39,40]. Additionally, efforts measuring metabolic changes using FET/PET imaging attempt to differentiate between mutational status or malignancy [41][42][43]. In our series, the correlation between FET/PET imaging and integrated diagnosis was low, with an accuracy of 23%. ...
Article
Full-text available
Simple Summary The preoperative risk estimation of non-enhancing suspected “low-grade” glioma (NEG) is key in determining the optimal timing of diagnosis and treatment to delay malignant progression and avoid undertreatment. The updated 2021 WHO classification brought new facets to glioma grading. Therefore, we sought to identify preoperative risk factors of malignancy in NEG by considering molecular criteria, including IDH mutation and CDKN2A/B deletion status. A total of 72 NEG patients were analyzed, and a high prevalence of malignant gliomas was detected considering both the traditional WHO grading (WHO grade 3 + 4) and the integrated molecular classification (IDHwt glioblastoma WHO grade 4 and IDHmut astrocytoma WHO grade 4). Easily determinable preoperative factors (age, T2/FLAIR mismatch sign, and SVZ involvement) were identified by uni- and multivariate analyses and incorporated into a score. The score estimates the probability of an NEG harboring a malignant glioma. Finally, the score was validated in a cohort of 40 NEG patients and proved to be a better prediction model than the Pignatti score or the T2/FLAIR mismatch sign. Abstract The preoperative grading of non-enhancing glioma (NEG) remains challenging. Herein, we analyzed clinical and magnetic resonance imaging (MRI) features to predict malignancy in NEG according to the 2021 WHO classification and developed a clinical score, facilitating risk estimation. A discovery cohort (2012–2017, n = 72) was analyzed for MRI and clinical features (T2/FLAIR mismatch sign, subventricular zone (SVZ) involvement, tumor volume, growth rate, age, Pignatti score, and symptoms). Despite a “low-grade” appearance on MRI, 81% of patients were classified as WHO grade 3 or 4. Malignancy was then stratified by: (1) WHO grade (WHO grade 2 vs. WHO grade 3 + 4) and (2) molecular criteria (IDHmut WHO grade 2 + 3 vs. IDHwt glioblastoma + IDHmut astrocytoma WHO grade 4). Age, Pignatti score, SVZ involvement, and T2/FLAIR mismatch sign predicted malignancy only when considering molecular criteria, including IDH mutation and CDKN2A/B deletion status. A multivariate regression confirmed age and T2/FLAIR mismatch sign as independent predictors (p = 0.0009; p = 0.011). A “risk estimation in non-enhancing glioma” (RENEG) score was derived and tested in a validation cohort (2018–2019, n = 40), yielding a higher predictive value than the Pignatti score or the T2/FLAIR mismatch sign (AUC of receiver operating characteristics = 0.89). The prevalence of malignant glioma was high in this series of NEGs, supporting an upfront diagnosis and treatment approach. A clinical score with robust test performance was developed that identifies patients at risk for malignancy.
... When combined with MRI, FET PET can allow for better evaluation of progression and response, as contrast is not affected by changes in the bloodbrain barrier (BBB) that can be caused by therapy [26,28,29]. These traits mean that FET PET has prognostic value in radiotherapy planning and results in better tumour tissue delineation for surgical planning [30][31][32]. FET PET's properties have led to interest in hybrid imaging systems, such as MR-PET. The combination of these imaging techniques improves tumour delineation compared to MRI, supplying metabolic and anatomical data invaluable to biopsy and surgical planning [33][34][35]. ...
Article
Full-text available
Glioblastoma multiforme (GBM) remains a challenging disease, as it is the most common and deadly brain tumour in adults and has no curative solution and an overall short survival time. This incurability and short survival time means that, despite its rarity (average incidence of 3.2 per 100,000 persons), there has been an increased effort to try to treat this disease. Standard of care in newly diagnosed glioblastoma is maximal tumour resection followed by initial concomitant radiotherapy and temozolomide (TMZ) and then further chemotherapy with TMZ. Imaging techniques are key not only to diagnose the extent of the affected tissue but also for surgery planning and even for intraoperative use. Eligible patients may combine TMZ with tumour treating fields (TTF) therapy, which delivers low-intensity and intermediate-frequency electric fields to arrest tumour growth. Nonetheless, the blood–brain barrier (BBB) and systemic side effects are obstacles to successful chemotherapy in GBM; thus, more targeted, custom therapies such as immunotherapy and nanotechnological drug delivery systems have been undergoing research with varying degrees of success. This review proposes an overview of the pathophysiology, possible treatments, and the most (not all) representative examples of the latest advancements.
... is also a part of the standard diagnostic protocol for glioblastoma and therefore does not require any extra procedures. If an MRI modality that is suitable for identifying the true extent of glioblastomas could be found, it would be a much more feasible examination to be used in clinical practice than 18 F-FET-PET (Muoio et al., 2018). ...
Article
Full-text available
Glioblastoma is the most frequent type of primary brain tumors. Despite the advanced therapy, most of the patients die within 2 years after the diagnosis. The tumor has a typical appearance on MRI: a central hypointensity surrounded by an inhomogeneous, ring-shaped contrast enhancement along its border. Too small to be recognized by MRI, detached individual tumor cells migrate along white matter fiber tracts several centimeters away from the edge of the tumor. Usually these cells are the source of tumor recurrence. If the infiltrated brain areas could be identified, longer survival time could be achieved through supratotal resection and individually planned radiation therapy. Probabilistic tractography is an advanced imaging method that can potentially be used to identify infiltrated pathways, thus the real extent of the glioblastoma. Our study consisted of twenty high grade glioma patients. Probabilistic tractography was started from the tumor. The location of tumor recurrence on follow-up MRI was considered as the primary infiltrated white matter tracts. The results of probabilistic tractography were evaluated at thirteen different thresholds. The overlap with the tumor recurrence of each threshold level was then defined to calculate the sensitivity and specificity. In the group level, sensitivity (81%) and specificity (90%) were the most reliable at 5% threshold level. There were two outliers in the study group, both with high specificity and very low sensitivity. According to our results, probabilistic tractography can help to define the true extent of the glioblastoma at the time of diagnosis with high sensitivity and specificity. Individually planned surgery and irradiation could provide a better chance of survival in these patients.
... Other studies confirmed these findings [57][58][59]. The 18 F-FET PET also seems to produce valuable information in the non-invasive determination of the molecular subtype, prognosis, and treatment (mainly for the determination of the biological tumor volume) and for the interpretation of early changes after treatment, when compared with MRI [60]. Since patients that do not benefit from treatment can possibly be identified in an early stage with 18 F-FET PET, treatment regimens can be changed more rapidly, reducing the treatment burden and optimizing individualized treatment regimens. ...
Article
Full-text available
Gliomas are the most frequent primary tumors of the brain. They can be divided into grade II-IV astrocytomas and grade II-III oligodendrogliomas, based on their histomolecular profile. The prognosis and treatment is highly dependent on grade and well-identified prognostic and/or predictive molecular markers. Multi-parametric MRI, including diffusion weighted imaging, perfusion, and MR spectroscopy, showed increasing value in the non-invasive characterization of specific molecular subsets of gliomas. Radiolabeled amino-acid analogues, such as 18F-FET, have also been proven valuable in glioma imaging. These tracers not only contribute in the diagnostic process by detecting areas of dedifferentiation in diffuse gliomas, but this technique is also valuable in the follow-up of gliomas, as it can differentiate pseudo-progression from real tumor progression. Since multi-parametric MRI and 18F-FET PET are complementary imaging techniques, there may be a synergistic role for PET-MRI imaging in the neuro-oncological imaging of primary brain tumors. This could be of value for both primary staging, as well as during treatment and follow-up.
... 18F-fluoro-ethyl-tyrosine (18F-FET) is a fluorinated amino acid used to detect brain tumours. 18F-FET is taken up into neoplastic cells due to their increased amino acid uptake through an L-type amino acid transport system, and it is not incorporated into proteins [25] (see Fig. 1). ...
Article
Full-text available
Introduction The aim of the narrative review was to analyse the applications of nuclear medicine (NM) techniques such as PET/CT with different tracers in combination with radiotherapy for the clinical management of glioblastoma patients. Materials and methods Key references were derived from a PubMed query. Hand searching and clinicaltrials.gov were also used. Results This paper contains a narrative report and a critical discussion of NM approaches in combination with radiotherapy in glioma patients. Conclusions NM can provide the Radiation Oncologist several aids that can be useful in the clinical management of glioblastoma patients. At the same, these results need to be validated in prospective and multicenter trials.
Thesis
Mit dieser Studie konnten wir durch retrospektive Beobachtungen zeigen, dass die strahlentherapeutische Behandlung von LGG im Verlauf relativ häufig zum Phänomen der PsPD führt (45,2 %). Darüber hinaus konnten wir in dieser Studie auch nachweisen, dass eine PsPD unabhängig von den drei Arten der analysierten Bestrahlungsmodalitäten aufgetreten ist (p=0,909). Die hohe Inzidenz (45,2 %) der PsPD in dieser und den zitierten Studien unterstreicht die Wichtigkeit der Unterscheidung eines echten PD von einem PsPD. Wir wollen mit dieser Studie dazu beitragen, dass zum einen unnötige Therapiewechsel oder Intensivierungstherapien bei einer PsPD vermieden werden, zum anderen, dass höhere Sicherheit bezüglich der Diagnose eines echten PD erreicht werden kann. Wenn klinisch tätige Ärzte darüber informiert werden, in welchen Lokalisationen bzw. unter welcher Strahlentherapieform ein erhöhtes Risiko für eine PsPD besteht, so können sie ein postradiogenes Wachstum mithilfe der kurzfristig ansteigenden PsPD-spezifischen Parameter im MRT eher einer PsPD als einem PD zuordnen. Auch die klinische Symptomatik muss im Verlauf bei Patienten mit LGG exakt analysiert werden. Die Follow-up Untersuchungen der asymptomatischen Patienten sind ein wichtiger Vergleichsparameter. Die aus dieser Studie gewonnenen Informationen können bei der Beurteilung hinsichtlich des Tumorprogress bei Patienten mit LGG und konsekutiver Radiotherapie helfen und möglicherweise auch Einfluss auf das Design künftiger pädiatrischer LGG Studien nehmen. Inwieweit andere Untersuchungsmodalitäten wie das FET-PET-CT eine exaktere Differenzierung zwischen PsPD und PD ermöglicht, müssen weitere Untersuchungen zeigen. Das FET-PET-CT scheint v.a. eine gute Ergänzung zu der reinen morphologischen Analyse darzustellen, da diese Methode auch die biologische Aktivität des Tumors erfassen kann. Während man mithilfe der MRT-Untersuchungen aufgrund dieser und vorherigen Studien mutmaßen kann, dass es sich bei kurzfristiger Zunahme der drei leitenden Parameter um eine PsPD handeln wird, könnte das FET-PET-CT eine noch größere Sicherheit hinsichtlich der Differenzierung PD versus PsPD geben, wobei einschränkend festzustellen ist, dass die diagnostischen Studien zur FET-PET rar sind.
Article
An 11-year-old boy who presented with headache and progressive right-sided weakness exhibited cortical swelling in the parafalcine area of both frontoparietal high convexity and splenium portion of corpus callosum on brain MRI. This suggested the possibility of encephalopathy, but required differential diagnosis from brain tumor. 18F-FET (O-(2-[18F]fluoroethyl)-l-tyrosine) PET/CT identified increased uptake along the parafalcine area of the frontoparietal lobes and the splenium portion of the corpus callosum. The relatively low target-to-background ratios were more indicative of inflammatory changes such as demyelinating disease. The patient recovered after empirical steroid and immunoglobulin treatment. Clinically, the patient was diagnosed with acute disseminated encephalomyelitis.
Article
Full-text available
Most high-grade gliomas (HGG) recur after initial multimodal therapy and re-irradiation (Re-RT) has been shown to be a valuable re-treatment option in selected patients. We evaluated the prognostic value of dynamic time-to-peak analysis and early static summation images in O-(2-¹⁸F-fluoroethyl)-l-tyrosine (¹⁸F-FET) PET for patients treated with Re-RT ± concomitant bevacizumab. We retrospectively analyzed 72 patients suffering from recurrent HGG with ¹⁸F-FET PET prior to Re-RT. PET analysis revealed the maximal tumor-to-background-ratio (TBRmax), the biological tumor volume, the number of PET-foci and pattern of time-activity-curves (TACs; increasing vs. decreasing). Furthermore, the novel PET parameters early TBRmax (at 5–15 min post-injection) and minimal time-to-peak (TTPmin) were evaluated. Additional analysis was performed for gender, age, KPS, O6-methylguanine-DNA methyltransferase methylation status, isocitrate dehydrogenase 1 mutational status, WHO grade and concomitant bevacizumab therapy. The influence of PET and clinical parameters on post-recurrence survival (PRS) was investigated. Shorter TTPmin was related to shorter PRS after Re-RT with 6 months for TTPmin < 12.5 min, 7 months for TTPmin 12.5–25 min and 11 months for TTPmin >25 min (p = 0.027). TTPmin had a significant impact on PRS both on univariate (p = 0.027; continuous) and multivariate analysis (p = 0.011; continuous). Other factors significantly related to PRS on multivariate analysis were increasing vs. decreasing TACs (p = 0.008) and Karnofsky Performance Score (p = 0.015; <70 vs. ≥70). Early TBRmax as well as the other conventional PET parameters were not significantly related to PRS on univariate analysis. Dynamic ¹⁸F-FET PET with TTPmin provides a high prognostic value for recurrent HGG prior to Re-RT, whereas early TBRmax does not. Dynamic ¹⁸F-FET PET using TTPmin might help to personalize Re-RT treatment regimens in future through voxelwise TTPmin analysis for dose painting purposes and PET-guided dose escalation.
Article
Full-text available
Rationale: Timely detection of pseudoprogression (PSP) is crucial for the management of patients with high-grade glioma (HGG) but remains difficult. Textural features of O-(2-[18F]fluoroethyl)-L-tyrosine positron emission tomography (FET-PET) mirror tumor uptake heterogeneity; some of them may be associated with tumor progression. Methods: Fourteen patients with HGG and suspected of PSP underwent FET-PET imaging. A set of 19 conventional and textural FET-PET features were evaluated and subjected to unsupervised consensus clustering. The final diagnosis of true progression vs. PSP was based on follow-up MRI using RANO criteria. Results: Three robust clusters have been identified based on 10 predominantly textural FET-PET features. None of the patients with PSP fell into cluster 2, which was associated with high values for textural FET-PET markers of uptake heterogeneity. Three out of 4 patients with PSP were assigned to cluster 3 that was largely associated with low values of textural FET-PET features. By comparison, tumor-to-normal brain ratio (TNRmax) at the optimal cutoff 2.1 was less predictive of PSP (negative predictive value 57% for detecting true progression, p=0.07 vs. 75% with cluster 3, p=0.04). Principal conclusions: Clustering based on textural O-(2-[18F]fluoroethyl)-L-tyrosine PET features may provide valuable information in assessing the elusive phenomenon of pseudoprogression.
Article
Full-text available
Background After focused high dose radiotherapy of brain metastases, differentiation between tumor recurrence and radiation-induced lesions by conventional MRI is challenging. This study investigates the usefulness of dynamic O-(2-18F-Fluoroethyl)-L-Tyrosine positron emission tomography (18F-FET PET) in patients with MRI-based suspicion of tumor recurrence after focused high dose radiotherapy of brain metastases. Methods Twenty-two patients with 34 brain metastases (median age 61.9 years) were included. Due to follow-up scan evaluations after repeated treatment in a subset of patients, a total of 50 lesions with MRI-based suspicion of tumor recurrence after focused high dose radiotherapy could be evaluated. 18F-FET PET analysis included the assessment of maximum and mean tumor-to-background ratio (TBRmax and TBRmean) and analysis of time-activity-curves (TAC; increasing vs. decreasing) including minimal time-to-peak (TTPmin). PET parameters were correlated with histological findings and radiological-clinical follow-up evaluation. ResultsTumor recurrence was found in 21/50 cases (15/21 verified by histology, 6/21 by radiological-clinical follow-up) and radiation-induced changes in 29/50 cases (5/29 verified by histology, 24/29 by radiological-clinical follow-up). Median clinical-radiological follow-up was 28.3 months (range 4.2–99.1 months). 18F-FET uptake was higher in tumor recurrence compared to radiation-induced changes (TBRmax 2.9 vs. 2.0, p < 0.001; TBRmean 2.2 vs. 1.7, p < 0.001). Receiver-operating-characteristic (ROC) curve analysis revealed optimal cut-off values of 2.15 for TBRmax and 1.95 for TBRmean (sensitivity 86 %, specificity 79 %). Increasing TACs and long TTPmin were associated with radiation-induced changes, decreasing TACs with tumor recurrence (p = 0.01). By combination of TBR and TACs, sensitivity and specificity could be increased to 93 and 84 %. Conclusions In patients with MRI-suspected tumor recurrence after focused high dose radiotherapy, 18F-FET PET has a high sensitivity and specificity for the differentiation of vital tumor tissue and radiation-induced lesions.
Article
Objective: The aim of this study was to explore the impact of automated hotspot detection on surgical planning of ¹⁸FET PET-guided stereotactic serial biopsy. Method: Imaging of ten patients with brain lesions detected by MRI and showing increased ¹⁸FET uptake on PET who were retrospectively and randomly assigned to compose the study. Stereotactic biopsy plans (PET-guided and MR-guided) were performed by two neurosurgeons for each patient, independently and blinded. For PET-guided plans, biopsy target was achieved by means of an automated hotspot detection system. MR-guided plans targeted contrast enhancement areas or hyperintense areas in T2-weighted sequences. FET uptake ratio (UR) was determined in the biopsy trajectory across the lesion. Highest UR (HUR) from both planning techniques was compared. Results: Each single HUR obtained through PET-guided technique was higher than correspondent values from MR-guided technique. Mean HUR of 2.41 (SE ± 0.23) for PET-guided plans and 1.85 (±0.16) for MR-guided plans were respectively obtained. This difference was statistically significant (p = 0.002). Conclusion: The use of an automated hotspot detection system was able to provide higher FET HUR along stereotactic biopsy trajectories in comparison to those from MR-guided plans. The use of specially designed computational tools may refine surgical planning by improving biopsy targeting.
Article
Purpose: To assess the utility of 18F-fluoroethyl-L-tyrosine (FET) positron emission tomography/magnetic resonance imaging (PET/MRI) in distinguishing recurrence from radionecrosis. Materials and Methods: Thirty-two patients (25 males, 7 females) of glioma who had already undergone surgery/chemoradiotherapy and had enhancing brain lesions suspicious of recurrence were evaluated using integrated 18F-FET PET/MRI, and followed up with histopathology or clinical follow-up and/or MRI/PET/MRI imaging. Manually drawn regions of interest over areas of maximal enhancement or FET uptake were used to calculate tumor to background ratios [TBRmax, TBRmean], choline: creatine ratio [Cho: Cr ratio], normalized relative cerebral blood volume [N rCBVmean] and apparent diffusion coefficient [ADCmean]. Correlations were evaluated using Pearson's coefficient. Accuracy of each parameter was calculated using independent t-test and receiver operator curve (ROC) analysis while utility of all four parameters together using multivariate analysis of variance (MANOVA) for differentiating recurrence vs. radionecrosis was evaluated. Positive histopathology and imaging/clinical follow up served as the gold standard. Results: Twenty-four of the 32 patients were diagnosed with recurrent disease and 8 with radiation necrosis. Significant correlations were observed between TBRmaxand N rCBVmean (ρ =0.503; P = 0.003), TBRmean, and N rCBVmean (ρ =0.414; P = 0.018), TBRmaxand ADCmean (ρ = −0.52; P = 0.002), and TBRmeanand ADCmean(ρ = −0.518; P = 0.002). TBRmax, TBRmean, ADCmean, Cho: Cr ratios, and N rCBVmeanwere significant in differentiating recurrence from radiation necrosis with an accuracy of 94.1%, 88.2%, 80.4%, 96.4%, and 89.9%, respectively. MANOVA indicated that combination of all parameters demonstrated better evaluation of recurrence vs. necrosis than any single parameter. The diagnostic accuracy, sensitivity, and specificity using all MRI parameters were 93.75%, 96%, and 85.7%, and using all FET PET/MRI parameters was 96.87%, 100%, and 85.7%, respectively. Conclusions: Synergetic effect of multiple MR parameters evaluated together in addition to FET PET uptake highlights the fact that integrated 18F-FET PET/MRI might have the potential to impact management of patients with glioma by timely and conclusive recognition of true recurrence from radiation necrosis.
Article
A combination of morphological imaging of the brain with microstructural and functional imaging provides a comprehensive overview of the properties of individual tissues. While diffusion weighted imaging provides information about tissue cellularity, spectroscopic imaging allows us to evaluate the integrity of neurons and possible anaerobic glycolysis during tumor hypoxia, in addition to the presence of accelerated synthesis or degradation of cellular membranes; on the other hand, PET metabolic imaging is used to evaluate major metabolic pathways, determining the overall extent of the tumor (¹⁸F-FET, ¹⁸F-FDOPA, ¹⁸F-FCH) or the degree of differentiation (¹⁸F-FDG, ¹⁸F-FLT, ¹⁸F-FDOPA and ¹⁸F-FET). Multi-parameter analysis of tissue characteristics and determination of the phenotype of the tumor tissue is a natural advantage of PET/MRI scanning. The disadvantages are higher cost and limited availability in all centers with neuro-oncology surgery. PET/MRI scanning of brain tumors is one of the most promising indications since the earliest experiments with integrated PET/MRI imaging systems, and along with hybrid imaging of neurodegenerative diseases, represent a new direction in the development of neuroradiology on the path towards comprehensive imaging at the molecular level.
Article
Background and purpose: To evaluate the patterns of failure following clinical introduction of amino-acid O-(2-[(18)F]fluoroethyl)-L-tyrosine (FET)-PET-guided target definition for radiotherapy (RT) of glioblastoma patients. Materials and methods: The first 66 consecutive patients with confirmed histology, scanned using FET-PET/CT and MRI were selected for evaluation. Chemo-radiotherapy was delivered to a volume based on both MRI and FET-PET (PETvol). The volume of recurrence (RV) was defined on MRI data collected at the time of progression according to RANO criteria. Results: Fifty patients were evaluable, with median follow-up of 45months. Central, in-field, marginal and distant recurrences were observed for 82%, 10%, 2%, and 6% of the patients, respectively. We found a volumetric overlap of 26%, 31% and 39% of the RV with the contrast-enhancing MR volume, PETvol and the composite MRPETvol, respectively. MGMT-methylation (p=0.03), larger PETvol (p<0.001), and less extensive surgery (p<0.001), were associated with larger PETvol overlap. Conclusion: The combined MRPETvol had a stronger association with the recurrence volume than either of the modalities alone. Larger overlap of PETvol and RV was observed for patients with MGMT-methylation, less extensive surgery, and large PETvol on the RT-planning scans.
Article
Background: Despite an increasing number of O-(2-(18)F-fluoroethyl)-L-tyrosine ((18)F-FET) PET studies in supratentorial gliomas, studies regarding the usefulness of (18)F-FET PET in brainstem and spinal cord gliomas to date remain scarce. Methods: Thirty-six (18)F-FET PET scans were performed in 29 patients with brainstem (n = 29 scans) or spinal cord glioma (n = 7 scans). In 32 of 36 PET scans, a dynamic acquisition was performed. Fifteen scans in 15 patients were performed to assess newly diagnosed lesions, and 21 scans were obtained during follow-up: for diagnosing tumor progression (n = 15 scans in 14 patients) as well as for treatment monitoring (n = 6 scans in 3 patients). Four patients underwent additional serial scans (range, 1-2), and 3 of these 4 patients were examined for more than one indication. Maximum and mean tumor/brain ratios (TBRmax/mean) of (18)F-FET uptake (20-40 min post injection) as well as kinetic (18)F-FET uptake parameters were determined. Final diagnoses were confirmed histologically (54%) or by clinical follow-up (46%). Results: In all newly diagnosed high-grade (n = 3 patients) and in 5 of 11 patients with low-grade gliomas, (18)F-FET uptake was increased (TBRmax ≥2.5 and/or TBRmean ≥1.9). In 2 patients with newly diagnosed gliomas without MR contrast enhancement, (18)F-FET PET nevertheless showed increased metabolism. At suspected progression, the combination of TBRs with kinetic (18)F-FET parameters correctly identified presence or absence of progressive disease in 9 of 11 patients (82%). Conclusions: This preliminary study suggests that (18)F-FET PET adds valuable diagnostic information in brainstem and spinal cord glioma, particularly when the diagnostic information derived from MRI is equivocal.
Article
Purpose18F-fluorethyltyrosine-(FET)-PET and MRI-based relative cerebral blood volume (rCBV) have both been used to characterize gliomas. Recently, inter-individual correlations between peak static FET-uptake and rCBV have been reported. Herein, we assess the local intra-lesional relation between FET-PET parameters and rCBV. Methods Thirty untreated glioma patients (27 high-grade) underwent simultaneous PET/MRI on a 3 T hybrid scanner obtaining structural and dynamic susceptibility contrast sequences. Static FET-uptake and dynamic FET-slope were correlated with rCBV within tumour hotspots across patients and intra-lesionally using a mixed-effects model to account for inter-individual variation. Furthermore, maximal congruency of tumour volumes defined by FET-uptake and rCBV was determined. ResultsWhile the inter-individual relationship between peak static FET-uptake and rCBV could be confirmed, our intra-lesional, voxel-wise analysis revealed significant positive correlations (median r = 0.374, p < 0.0001). Similarly, significant inter- and intra-individual correlations were observed between FET-slope and rCBV. However, rCBV explained only 12% of the static and 5% of the dynamic FET-PET variance and maximal overlap of respective tumour volumes was 37% on average. Conclusions Our results show that the relation between peak values of MR-based rCBV and static FET-uptake can also be observed intra-individually on a voxel basis and also applies to a dynamic FET parameter, possibly determining hotspots of higher biological malignancy. However, just a small part of the FET-PET signal variance is explained by rCBV and tumour volumes determined by the two modalities showed only moderate overlap. These findings indicate that FET-PET and MR-based rCBV provide both congruent and complimentary information on glioma biology.
Article
Objectives We investigated the potential of textural feature analysis of O-(2-[18F]fluoroethyl)-L-tyrosine (18F-FET) PET to differentiate radiation injury from brain metastasis recurrence. Methods Forty-seven patients with contrast-enhancing brain lesions (n = 54) on MRI after radiotherapy of brain metastases underwent dynamic 18F-FET PET. Tumour-to-brain ratios (TBRs) of 18F-FET uptake and 62 textural parameters were determined on summed images 20-40 min post-injection. Tracer uptake kinetics, i.e., time-to-peak (TTP) and patterns of time-activity curves (TAC) were evaluated on dynamic PET data from 0-50 min post-injection. Diagnostic accuracy of investigated parameters and combinations thereof to discriminate between brain metastasis recurrence and radiation injury was compared. ResultsDiagnostic accuracy increased from 81 % for TBRmean alone to 85 % when combined with the textural parameter Coarseness or Short-zone emphasis. The accuracy of TBRmax alone was 83 % and increased to 85 % after combination with the textural parameters Coarseness, Short-zone emphasis, or Correlation. Analysis of TACs resulted in an accuracy of 70 % for kinetic pattern alone and increased to 83 % when combined with TBRmax. Conclusions Textural feature analysis in combination with TBRs may have the potential to increase diagnostic accuracy for discrimination between brain metastasis recurrence and radiation injury, without the need for dynamic 18F-FET PET scans. Key points• Textural feature analysis provides quantitative information about tumour heterogeneity• Textural features help improve discrimination between brain metastasis recurrence and radiation injury• Textural features might be helpful to further understand tumour heterogeneity• Analysis does not require a more time consuming dynamic PET acquisition