Skip to main content
Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging 8/2011

Open Access 01.08.2011 | Original Article

Can FDG PET predict radiation treatment outcome in head and neck cancer? Results of a prospective study

verfasst von: Dominic A. X. Schinagl, Paul N. Span, Wim J. Oyen, Johannes H. A. M. Kaanders

Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging | Ausgabe 8/2011

Abstract

Purpose

In head and neck cancer (HNC) various treatment strategies have been developed to improve outcome, but selecting patients for these intensified treatments remains difficult. Therefore, identification of novel pretreatment assays to predict outcome is of interest. In HNC there are indications that pretreatment tumour 18F-fluorodeoxyglucose (FDG) uptake may be an independent prognostic factor. The aim of this study was to assess the prognostic value of FDG uptake and CT-based and FDG PET-based primary tumour volume measurements in patients with HNC treated with (chemo)radiotherapy.

Methods

A total of 77 patients with stage II–IV HNC who were eligible for definitive (chemo)radiotherapy underwent coregistered pretreatment CT and FDG PET. The gross tumour volume of the primary tumour was determined on the CT (GTVCT) and FDG PET scans. Five PET segmentation methods were applied: interpreting FDG PET visually (PETVIS), applying an isocontour at a standardized uptake value (SUV) of 2.5 (PET2.5), using fixed thresholds of 40% and 50% (PET40%, PET50%) of the maximum intratumoral FDG activity (SUVMAX) and applying an adaptive threshold based on the signal-to-background (PETSBR). Mean FDG uptake for each PET-based volume was recorded (SUVmean). Subsequently, to determine the metabolic volume, the integrated SUV was calculated as the product of PET-based volume and SUVmean. All these variables were analysed as potential predictors of local control (LC), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), disease-free survival (DFS) and overall survival (OS).

Results

In oral cavity/oropharynx tumours PETVIS was the only volume-based method able to predict LC. Both PETVIS and GTVCT were able to predict DMFS, DFS and OS in these subsites. Integrated SUVs were associated with LC, DMFS, DFS and OS, while SUVmean and SUVMAX were not. In hypopharyngeal/laryngeal tumours none of the variables was associated with outcome.

Conclusion

There is no role yet for pretreatment FDG PET as a predictor of (chemo)radiotherapy outcome in HNC in daily routine. However, this potential application needs further exploration, focusing both on FDG PET-based primary tumour volume, integrated SUV and SUVMAX of the primary tumour.

Introduction

In head and neck cancer various treatment strategies have been developed to improve outcome. However, it remains difficult to select patients for these intensified treatments despite careful evaluation of clinical factors such as tumour size/stage, lymph node involvement and anatomic subsite. Therefore, identification of novel pretreatment factors that potentially predict treatment response and long-term outcome is of great interest [1]. The development of molecular imaging techniques, such as PET, allows the noninvasive study of the pathophysiology of cancers.
In head and neck cancer there are indications that pretreatment tumour 18F-fluorodeoxyglucose (FDG) uptake may be an independent prognostic factor [1]. Many research groups have studied the incorporation of FDG PET into radiation treatment planning, and several ways of using PET data have been described. Visual interpretation is the most commonly used method [25]. This method, however, is susceptible to variations due to the window level settings of the images and is highly operator-dependent. Therefore, more objective methods have been explored. Examples are isocontouring based on a standardized uptake value (SUV) of 2.5 around the tumour [3, 68], a fixed threshold of the maximum signal intensity [913], or a threshold which is adaptive to the signal to background ratio (SBR) [3, 14]. We recently demonstrated that FDG PET may have important consequences for the definition of the gross tumour volume (GTV) of the primary tumour in head and neck cancer, and that the choice of the PET segmentation tool is not trivial [15]. The aim of this study was to assess the prognostic value of the determination of primary tumour volume from CT and FDG PET scans, and various ways of quantifying FDG uptake in patients with head and neck cancer treated with (chemo)radiotherapy, and to provide an overview of the available literature.

Material and methods

Patients

A total of 77 patients (58 men and 19 women; median age 61 years, range 43–86 years) with stage II–IV squamous cell carcinoma of the head and neck area, eligible for primary curative radiotherapy, were prospectively enrolled from June 2003 until July 2006. FDG PET was performed only for research purposes, and did not influence treatment. The tumour characteristics are summarized in Table 1. No information on human papillomavirus relatedness can be provided. The study was approved by the Ethics Committee of the Radboud University Nijmegen Medical Centre and all patients provided informed consent.
Table 1
Tumour characteristics of 77 patients
Tumour characteristic
No. of patients
Site
 
 Oral cavity
6
 Oropharynx
30
 Hypopharynx
9
 Larynx
32
T stage
 
 T1
1
 T2
15
 T3
39
 T4
22
N stage
 
 N0
21
 N1
10
 N2a
0
 N2b
17
 N2c
28
 N3
1
Histological grade
 
 1
4
 2
37
 3
33
 Unknown
3
Total
77

Treatment

All patients were discussed in a multidisciplinary conference for tumour classification and treatment recommendations. Our protocol recommended treating primary tumour and metastatic lymph nodes to a dose of 68–70 Gy This was combined with concomitant weekly intravenous cisplatinum 40 mg/m2 for large unresectable tumours. Elective lymph node regions were treated to 44 Gy.

Image acquisition

Before treatment, a CT scan and an FDG PET scan were acquired in radiation treatment position with the patient immobilized using a custom-made rigid mask covering the head, neck and shoulders. Maximum reproducibility in positioning was ensured by the use of additional support systems: a flat scanning bed, customized head support cushion, intraoral mould when indicated, standard cushion supporting the knees, and laser positioning system as previously described [15]. CT scans were acquired using a multislice spiral CT scanner (Philips AcQsim; Philips, Cleveland, OH). Scanning parameters were 130 kV, 120 mAs, slice distance and slice thickness 3 mm, scanning the head and neck area, with intravenous contrast agent. FDG PET scans were acquired using a full-ring dedicated PET scanner (Siemens ECAT Exact 47; Siemens/CTI, Knoxville, TN). Patients with diabetes mellitus were not excluded. However, glucose levels had to be appropriately regulated (glucose level at time of FDG injection <10 mmol/l, no insulin administration before FDG injection). A 3-D emission scan of the head and neck area and a 2-D 68GE-based transmission scan for attenuation correction were acquired 60 min (median±SD 64±11.4 min) after intravenous injection of 250 MBq FDG (Covidien, Petten, The Netherlands). The acquisition time per bed position was 5 min for emission and 3 min for the Ge-based transmission scan, resulting in a total scanning time of 16 min for the two bed positions. Image reconstruction has been described in detail previously [16].
Three-dimensional surface models were automatically derived from both the CT and PET images. These models were anatomically coregistered using an operator-independent iterative closest point algorithm, with an average registration error of 2.0 mm at the centre of the planning area as described previously [17]. SUV was defined as the voxel value of detected activity multiplied by the weight of the patient divided by the activity at the beginning of the scan.
The CT and the two PET datasets were transferred via DICOM to a Pinnacle3 treatment planning system (Philips Medical Systems, Andover, MA) for target volume definition.

Target volume definition

The primary tumour was delineated on CT and FDG PET images by two experienced radiation oncologists in consensus. The volume of the metastatic lymph nodes was not included. The role of FDG PET in the delineation of metastatic lymph nodes has been analysed previously [18].
On CT images, the GTV (GTVCT) was delineated manually according to current clinical protocols using information gathered from physical examination, available diagnostic work-up imaging (CT and/or MRI, examination under general anaesthesia) and the CT scan in treatment position. When the radiation oncologists were drawing the GTVCT contours, the FDG PET images were blinded.
Five PET-based volumes were obtained using different delineation approaches. The volumes were delineated visually (PETVIS) by contouring the FDG activity that was clearly above normal background activity. Locations with increased FDG uptake were classified as malignant in consensus with an experienced nuclear medicine physician. The other (threshold-based) volumes were obtained using in-house developed software scripts for the Pinnacle3 treatment planning system. Volumes were delineated by applying an isocontour of SUV = 2.5 (PET2.5) around the tumour. Volumes were delineated using two fixed percentage thresholds of 40% (PET40%) and 50% (PET50%) of the maximum signal intensity in the primary tumour (SUVMAX). Finally, volumes were delineated using an adaptive threshold based on the SBR (PETSBR), as developed at Université St. Luc in Brussels, Belgium [14]. Calibration and implementation of the PETSBR method have been described in detail previously [15]. Results obtained by automated delineation algorithms were checked visually before acceptance. A delineation was considered unsuccessful if the resulting volume included significant volumes of tissue that were clearly normal on visual interpretation.
The mean FDG uptake of each PET-based volume was recorded (SUVmeanVIS, SUVmean2.5, SUVmean40%, SUVmean50%, SUVmeanSBR). This was multiplied by the corresponding volume resulting in the integrated SUV (iSUVVIS, iSUV2.5, iSUV40%, iSUV50%, iSUVSBR).

Treatment outcome analysis

Follow-up visits included history, inspection of the upper aerodigestive tract and palpation of the neck. Local and regional recurrences were proven by histology and cytology, respectively. Distant metastases were identified by either pathologically or radiologically.

Statistics

All statistical analyses were performed using SPSS version 16.0 (SPSS, Chicago, IL). The significances of differences between two categories were established using t-tests or Mann-Whitney U testing, when appropriate. The normality of distributions were assessed using Kolmogorov-Smirnov tests. Variables were entered as continuous variables in Cox regression analyses to avoid the need to establish a cut-off value for local control (LC), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), disease-free survival (DFS) and overall survival (OS). A p < 0.05 was a priori considered as statistically significant.

Results

Tumour volume measurements

For CT-based primary tumour volume measurements, 77 datasets were available. PETVIS was generated for all 77 patients; the PETSBR segmentation tool resulted in unsuccessful volume definition in two patients. A delineation was considered unsuccessful if the resulting GTV included significant volumes of tissue that were clearly normal on visual interpretation. This was observed in four patients for both PET40% and PET50%, two of whom also had an unsatisfactory PETSBR. The PET2.5 segmentation tool was unsuccessful in 35 patients, including the four patients mentioned. As a consequence, this latter method was not evaluated further. All unsuccessful volume definitions were largely over-sized, being at least 300 cm3 and clearly incorporated benign tissue. An unsuccessful delineation did not correlate with specific tumour subsite or T stage. An example of an inadequate PET2.5 is shown in Fig. 1. The mean absolute tumour volume for the various methods were 22.7, 21.5, 16.4, 10.5 and 11.2 cm3 for GTVCT, PETVIS, PET40%, PET50% and PETSBR, respectively. GTVCT and PETVIS yielded similar mean absolute volumes, but the threshold-based methods (PET40%, PET50% and PETSBR) yielded volumes that were all smaller than GTVCT (p ≤ 0.0001 for all comparisons). Overlap and mismatch analyses performed in order to evaluate the location of the acquired volumes showed that in 64%, 59%, 29% and 31% of the PETVIS, PET40%, PET50% and PETSBR volumes, respectively, more than 20% of the volume was located outside the GTVCT domain.

Treatment and treatment outcome

The median primary tumour radiation dose was 68 Gy (range 64–72 Gy). Three patients were not treated; one died just prior to radiotherapy, another refused primary radiotherapy, and the third developed distant metastases prior to radiotherapy. After a median follow-up of 46 months (range 2.5–76 months), LC, RRFS, DMFS, DFS and OS at 2 years were 84%, 95%, 86%, 73% and 77%, respectively. Follow-up was at least 24 months or until the patient’s death. After primary treatment, five patients did not achieve complete remission. These patients did not have significantly different CT- or PET-based tumour volumes from the patients who did achieve complete remission. No recurrences were seen in the areas treated with an elective dose.

Prognostic value of CT and PET

Primary tumour volume (PET- or CT-based), SUVmean, SUVMAX and iSUV were not able to predict the likelihood of complete remission. The CT- and PET-based tumour volumes of the patients who did achieve complete remission (n = 69) are shown in Fig. 2. There was a significant difference in the volumes of oral cavity and oropharyngeal tumours as compared to laryngeal and hypopharyngeal tumours (p ≤ 0.004, Mann-Whitney). The values of SUVMAX for oral cavity/oropharyngeal tumours and laryngeal/hypopharyngeal tumours were 9.7 and 10.0, respectively. We analysed LC, RRFS, DMFS, DFS and OS in the 69 patients who achieved complete remission after primary treatment using primary tumour volume (PET- or CT-based), SUVmean, SUVMAX and iSUV as continuous variables in Cox regression survival analyses.
In hypopharyngeal and laryngeal tumours, none of the CT or PET parameters was associated with any of the outcome-related endpoints. SUVMAX and SUVmean also had no prognostic value in oral cavity and oropharyngeal tumours. The other results for oral cavity and oropharyngeal tumours are presented in Table 2. In these head and neck subsites, PETVIS was able to predict LC, whereas the other volume-based methods were not. Both PETVIS and GTVCT were able to predict DMFS, DFS and OS. Furthermore, all iSUV methods were able to predict LC, DMFS, DFS, and OS, albeit sometimes with borderline significance (p-values between 0.051 and 0.055). Figure 3 shows individual data points of GTVCT and PETVIS in relation to LC and DFS of oral cavity/oropharyngeal tumours with a follow-up of at least 24 months. Although the mean values differed significantly, Fig. 3 also shows that there was a large overlap in the volume range between patients with and without recurrence or death, indicating that the discriminative power of GTVCT and PETVIS is limited.
Table 2
Primary tumour volume (PET- or CT-based) and PET-based iSUV as variables in treatment outcome prediction in patients with oral cavity and oropharynx tumours who achieved complete remission (n = 31) after definitive (chemo)radiotherapy. Variables were assessed using Cox regression analysis. The values shown are p-values
Outcome
GTVCT
PETVIS
PET40%
PET50%
PETSBR
iSUVVIS
iSUV40%
iSUV50%
iSUVSBR
LC
>0.1
0.031
>0.1
>0.1
>0.1
0.021
0.025
0.039
0.033
RRFS
>0.1
>0.1
>0.1
>0.1
>0.1
>0.1
>0.1
>0.1
>0.1
DMFS
0.003
0.046
0.080
0.064
>0.1
0.055
0.023
0.023
0.024
DFS
0.024
0.016
>0.1
>0.1
>0.1
0.033
0.041
0.054
0.051
OS
0.018
0.023
>0.1
>0.1
>0.1
0.026
0.038
0.052
0.040

Discussion

In this study we assessed the prognostic value of CT- and FDG PET-based primary tumour volume measurements, mean FDG uptake (SUVmean) and maximum FDG uptake (SUVMAX), and iSUV in a large cohort of patients with head-and-neck cancer treated with (chemo)radiotherapy.
Interestingly, PETVIS was able to predict LC of oral cavity and oropharyngeal tumours, but GTVCT was not, while the mean PETVIS and GTVCT volumes were similar. Other studies have confirmed the lack of prognostic potential of CT-based primary tumour volume in oral cavity and oropharyngeal tumours [33, 34]. Our observation that PETVIS is associated with LC is novel. It remains questionable, however, if visual assessment can be a reliable prognostic tool given the operator-dependent nature of this method. Both GTVCT and PETVIS were able to predict DMFS, DFS and OS in these subsites. For CT-based primary tumour volume this was also observed by Chao et al. in 31 patients with oropharyngeal cancer treated with definitive (chemo)radiotherapy [35]. Apparently, in oropharynx tumours local radiotherapy response does not depend so much on the primary tumour volume, but possibly more on the biological characteristics of the tumour [36]. On the other hand, these results do suggest that metastatic potential is associated with the primary tumour volume in this head and neck subsite. One other study of 59 patients with stage III–IV head and neck cancer treated with definitive (chemo)radiotherapy found a correlation between PET-based primary tumour volume, using the PET2.5 method, and PFS [28]. After further analyses the study also showed that a volume ≥9.3 cm3 was associated with a decreased OS.
All the iSUV methods (the product of the PET-based primary tumour volume and the SUVmean within that volume, reflecting the metabolic volumes) were able to predict LC, DMFS, DFS and OS in oral cavity and oropharynx tumours, albeit sometimes with borderline significance. iSUV is a new variable fully representing the total metabolic activity within a predefined tumour volume. La et al. also found a correlation between iSUV and treatment outcome, albeit based on cumulative volumes of both the primary tumour and the PET-avid lymph nodes [27]. However, they hypothesized that the effect was due to the volume and not the product of volume and SUVmean. In contrast, our data indicate that of all the PET-based volume measurements, only PETVIS had a predictive value, while this was the case for practically all the iSUV methods. This suggests that the product of volume and SUVmean provides a more robust parameter which could possibly be a surrogate for both tumour aggressiveness and the total cancer cell mass.
In hypopharyngeal and laryngeal tumours we found no association between GTVCT or PETVIS and treatment outcome, whereas several studies have demonstrated the prognostic value of CT-determined tumour volume for outcome after definitive radiation therapy for these subsites as well as for nasopharyngeal cancer [37]. We do not have a solid explanation for this observation, except for the fact that we obtained high tumour control rates (LC at 2 years of 86%) compared to several other studies, and consequently relatively few events which would reduce the discriminative power of any pretreatment test. None of the three semiquantitative methods for PET-based tumour volume calculation (PET40%, PET50% and PETSBR) showed an association with outcome in any of the head and neck subsites. It should be noted that all three semiquantitative methods produced significantly smaller variability. This may also reduce discriminative power.
As the absolute volumes of FDG PET-based tumour sometimes partly located outside the GTVCT domain were small, it was not possible to determine whether the exact origin of a recurrence lay located outside the GTVCT domain, but within the FDG PET-based tumour volume.
In our cohort the SUVMAX of the primary tumour was not able to predict radiation treatment outcome. Table 3 summarizes the results of a literature search for studies examining the role of pretreatment FDG PET SUVMAX in patients with head and neck cancer treated with definitive (chemo)radiotherapy in predicting outcome. Of 15 studies identified, 8 showed that SUVMAX could possibly play a role in predicting radiation treatment response [1, 1925] and 7 showed that it does not [2632]. These inconsistencies could be a result of the heterogeneity of treatment modalities, the heterogeneity of tumour sites, the use of several endpoints (i.e. LC, LRF, DFS or OS), the use of various SUVMAX cut-off values, and the use of either the SUVMAX of the primary tumour or the SUVMAX of a metastatic lymph node. It is important to note that of the eight studies demonstrating an association between SUVMAX and outcome, six included substantial numbers of patients who were treated with surgery. Overall, of the 408 patients included in these six studies, 227 (55%) underwent primary surgery. In fact, the study by Brun et al. is the only one indicating that SUVMAX is a prognostic factor in a population treated with definitive (chemo)radiotherapy alone, and using only the SUVMAX of the primary tumour, finding that DFS and OS were worse when SUVMAX was >9.0 [19]. Thus, based on this overview of the literature, an unequivocal conclusion about the predictive role of pretreatment FDG PET SUVMAX in patients with head and neck cancer treated with definitive (chemo)radiotherapy cannot yet be drawn. Possibly a studies of larger cohorts of patients with homogeneous tumours and treatment characteristics stratified for the various subsites would be able to establish a role for a SUVMAX cut-off value in order to investigate future treatment individualization. Ideally these studies should use the same type of treatment and the same definition of treatment outcome.
Table 3
Summary of studies on treatment outcome prediction using SUVMAX from pretreatment FDG PET of patients with head and neck cancer treated with definitive (chemo)radiotherapy
Reference
No. of patients
Tumour site
Variable
Prediction
Treatment results
21
41
Nasopharynx (n = 41)
SUVMAX primary tumour and/or metastatic lymph node
DFS worse when SUVMAX >8.0
3 years DFS 74.3%
22
60a
Oral cavity/oropharynx (n = 44)
SUVMAX primary tumour and/or metastatic lymph node
DFS and OS worse when SUVMAX ≥9.0
If SUVMAX ≥9.0 then 2 years DFS 37%; if <9.0 then 2 years DFS 76%
Hypopharynx/Larynx (n = 16)
30
45
Nasopharynx (n = 16)
SUVMAX primary tumour
DFS not correlated with SUVMAX
If SUVMAX ≥5.5 then 2 years DFS 48%; if <5.5 then 2 years DFS 76%
Oropharynx (n = 20)
Hypopharynx (n = 3)
Others (n = 6)
19
47
Nasopharynx (n = 6)
SUVMAX primary tumour
DFS and OS worse when SUVMAX >9.0
LC 78% (‘during follow-up time’)
Oral cavity/oropharynx (n = 30)
Hypopharynx/larynx (n = 10)
Maxilla (n = 1)
25
54b
Oral cavity/oropharynx (n = 34)
SUVMAX primary tumour
LC and DFS worse when SUVMAX ≥9.0
If SUVMAX ≥9.0 then 2 years LC 73%; <9.0 then 2 years LC 96%; ≥9.0 then 2 years DFS 69%; <9.0 then 2 years DFS 93%
Hypopharynx/larynx (n = 20)
1
120c
Oral cavity/oropharynx (n = 78)
SUVMAX primary tumour or metastatic lymph node
LC and DFS worse when SUVMAX >4.8
4 years LC 75%; 4 years DFS 59%
Hypopharynx/larynx (n = 39)
Unknown (n = 3)
31
12
Oral cavity/oropharynx (n = 6)
SUVMAX primary tumour or metastatic lymph node
LC not correlated with SUVMAX
LC 58% (‘during follow-up time’)
Hypopharynx/larynx (n = 5)
Unknown (n = 1)
24
79d
Hypopharynx/larynx (n = 79)
SUVMAX primary tumour
LC and DFS worse when SUVMAX >8.0
3 years LC 79%; 3 years DFS 50%
20
58e
Nasopharynx (n = 1)
SUVMAX primary tumour
OS worse when SUVMAX >10.0
No LC or DFS data provided
Oral cavity/oropharynx (n = 55)
Hypopharynx (n = 1)
Maxilla (n = 1)
23
37f
Nasopharynx (n = 5)
SUVMAX primary tumour
OS worse when SUVMAX >9
No LC or DFS data provided
Oral cavity/oropharynx (n = 16)
Hypopharynx/larynx (n = 15)
Parotid gland (n = 2)
26
82
Nasopharynx (n = 63)
SUVMAX primary tumour
DFS not correlated with SUVMAX
DFS 78% (‘after mean follow-up 35 months’)
Oropharynx (n = 13)
Hypopharynx (n = 6)
29
61
Nasopharynx (n = 2)
SUVMAX primary tumour or metastatic lymph node
LRF not correlated with SUVMAX
2 years LRF 17%
Oral cavity/oropharynx (n = 46)
Hypopharynx/larynx (n = 9)
Unknown (n = 4)
28
59
Oropharynx (n = 13)
SUVMAX primary tumour
PFS and OS not correlated to SUVMAX
No LC or DFS data provided
Hypopharynx/larynx (n = 46)
27
85
Nasopharynx (n = 22)
SUVMAX primary tumour or metastatic lymph node
DFS and OS not correlated with SUVMAX
2 years DFS 70%; 2 years OS 78%
Oral cavity/oropharynx (n = 49)
Hypopharynx/larynx (n = 12)
Unknown (n = 2)
32
42
Nasopharynx (n = 3)
SUVMAX primary tumour or metastatic lymph node
DFS and OS not correlated with SUVMAX
2 years DFS 71%; 2 years OS 83%
Oral cavity/oropharynx (n = 27)
Hypopharynx/larynx (n = 8)
Unknown (n = 4)
Current study
74
Oral cavity/oropharynx (n = 36)
SUVMAX primary tumour
LC, DFS and OS not correlated with SUVMAX
2 years LC 84%; 2 years DFS 73%
Hypopharynx/larynx (n = 38)
a19 patients definitive (chemo)radiotherapy, 41 patients surgery and (chemo)radiotherapy.
b27 patients definitive (chemo)radiotherapy, 17 patients surgery and radiotherapy, 8 patients surgery, 1 patient chemotherapy, 1 patient not reported.
c73 patients definitive (chemo)radiotherapy, 31 patients surgery and radiotherapy, 16 patients surgery.
d37 patients definitive (chemo)radiotherapy, 34 patients surgery and radiotherapy, 6 patients surgery, 2 patients (chemo)radiotherapy and surgery.
e5 patients definitive radiotherapy, 40 patients surgery and radiotherapy, 13 patients surgery.
f16 patients definitive radiotherapy, 2 patients surgery and radiotherapy, 19 patients radiotherapy and surgery.
Using pretreatment primary tumour volume based on FDG PET is appealing, and has not yet been extensively reported. In the current study, PETVIS proved to be the only PET-based volume able to predict treatment outcome, and only in the oral cavity and oropharyngeal tumours. It should be noted that the discriminative potential of PETVIS may be limited because of the large overlap between data points of patients with and without recurrence. The volumes generated by semiautomated PET segmentation methods were not useful for outcome prediction.
Thorwarth et al. demonstrated that cumulative FDG PET-based volumes of both the primary tumour and the PET-avid lymph nodes could not predict treatment outcome in a small series of patients with head and neck cancer treated with definitive (chemo)radiotherapy [31]. They generated the PET-based volume by encompassing all voxels showing a higher intensity than 40% of the maximum value. La et al. showed a correlation between DFS and OS of 85 patients with head and neck cancer treated with definitive (chemo)radiotherapy and the FDG PET-based cumulative volumes of both the primary tumour and the PET-avid lymph nodes [27]. They generated the PET-based volume by encompassing all voxels showing a higher intensity than 50% of the maximum value. Recently, Chung et al. showed a correlation between the DFS of 82 patients with pharyngeal cancer treated with definitive (chemo)radiotherapy and the FDG PET-based cumulative volumes of both the primary tumour and the PET-avid lymph nodes [26]. They generated the PET-based volume by encompassing all voxels showing an SUV of ≥2.5, and this was significant prognostic factor for DFS, whereas stage, histological grade and SUVMAX were not. In our cohort, the PET2.5 segmentation method resulted in an unsuccessful delineation in 35 patients, and factors that might explain this finding have been addressed in a previous report [15].
The use of a molecular imaging modality such as FDG PET to identify a robust variable on which prediction of treatment response and long-term outcome can be based remains attractive. Thus far, there is no role for pretreatment FDG PET as a predictor of outcome in head and neck cancer in daily routine, given the inconsistencies between studies and the low levels of evidence. However, this potential application of FDG PET needs further exploration, focusing both on FDG PET-based primary tumour volume and on iSUV and SUVMAX of the primary tumour. Preferably these questions should be incorporated in prospective phase III trials with strict criteria on treatment and outcome parameters. Other research questions are worth considering such as adding the data of a repeat FDG PET scan during treatment to the data acquired by a pretreatment FDG PET scan, and the use of different PET tracers such as 18F-fluoromisonidazole and 3′-deoxy-3′-18F-fluorothymidine, to image hypoxia and tumour cell proliferation, respectively, which are well-known tumour characteristics relevant to radiation response [38].

Conclusion

There are three major findings of this study. First, in oral cavity and oropharyngeal tumours PETVIS was the only volume-based method able to predict LC. Both PETVIS and GTVCT were associated with DMFS, DFS and OS in these subsites. Second, in oral cavity and oropharyngeal tumours the volume- and SUV-derived parameters iSUVVIS, iSUV40%, iSUV50%, iSUVSBR were consistently associated with LC, DMFS, DFS and OS, while SUVmean and SUVMAX were not. Third, in hypopharyngeal and laryngeal tumours, none of the CT and PET parameters was correlated with treatment outcome.
Given the inconsistencies between studies and low level of evidence thus far, there is no role yet for pretreatment FDG PET as a predictor of outcome in head and neck cancer in daily routine. Due to the heterogeneous nature of head and neck cancers, the difficulty in obtaining a large number of patients, and the variation in results, one has to be careful interpreting the results from our and similar studies, as they are based on a relatively low number of events. However, this potential application of FDG PET needs further exploration, focusing both on FDG PET-based primary tumour volume and on iSUV and SUVMAX of the primary tumour. Preferably these questions should be incorporated in prospective phase III trials with strict criteria on treatment and outcome parameters.

Conflicts of interest

None.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Allal AS, Slosman DO, Kebdani T, Allaoua M, Lehmann W, Dulguerov P. Prediction of outcome in head-and-neck cancer patients using the standardized uptake value of 2-[18F]fluoro-2-deoxy-D-glucose. Int J Radiat Oncol Biol Phys. 2004;59:1295–300.PubMedCrossRef Allal AS, Slosman DO, Kebdani T, Allaoua M, Lehmann W, Dulguerov P. Prediction of outcome in head-and-neck cancer patients using the standardized uptake value of 2-[18F]fluoro-2-deoxy-D-glucose. Int J Radiat Oncol Biol Phys. 2004;59:1295–300.PubMedCrossRef
2.
Zurück zum Zitat Heron DE, Andrade RS, Flickinger J, Johnson J, Agarwala SS, Wu A, et al. Hybrid PET-CT simulation for radiation treatment planning in head-and-neck cancers: a brief technical report. Int J Radiat Oncol Biol Phys. 2004;60:1419–24.PubMedCrossRef Heron DE, Andrade RS, Flickinger J, Johnson J, Agarwala SS, Wu A, et al. Hybrid PET-CT simulation for radiation treatment planning in head-and-neck cancers: a brief technical report. Int J Radiat Oncol Biol Phys. 2004;60:1419–24.PubMedCrossRef
3.
Zurück zum Zitat Nestle U, Kremp S, Schaefer-Schuler A, Sebastian-Welsch C, Hellwig D, Rübe C, et al. Comparison of different methods for delineation of 18F-FDG PET-positive tissue for target volume definition in radiotherapy of patients with non-small cell lung cancer. J Nucl Med. 2005;46:1342–8.PubMed Nestle U, Kremp S, Schaefer-Schuler A, Sebastian-Welsch C, Hellwig D, Rübe C, et al. Comparison of different methods for delineation of 18F-FDG PET-positive tissue for target volume definition in radiotherapy of patients with non-small cell lung cancer. J Nucl Med. 2005;46:1342–8.PubMed
4.
Zurück zum Zitat Nishioka T, Shiga T, Shirato H, Tsukamoto E, Tsuchiya K, Kato T, et al. Image fusion between 18FDG-PET and MRI/CT for radiotherapy planning for oropharyngeal and nasopharyngeal carcinomas. Int J Radiat Oncol Biol Phys. 2002;53:1051–7.PubMedCrossRef Nishioka T, Shiga T, Shirato H, Tsukamoto E, Tsuchiya K, Kato T, et al. Image fusion between 18FDG-PET and MRI/CT for radiotherapy planning for oropharyngeal and nasopharyngeal carcinomas. Int J Radiat Oncol Biol Phys. 2002;53:1051–7.PubMedCrossRef
5.
Zurück zum Zitat Riegel AC, Berson AM, Destian S, Ng T, Tena LB, Mitnick RJ, et al. Variability of gross tumor volume delineation in non-small-lung cancer. Int J Radiat Oncol Biol Phys. 2006;65:726–32.PubMedCrossRef Riegel AC, Berson AM, Destian S, Ng T, Tena LB, Mitnick RJ, et al. Variability of gross tumor volume delineation in non-small-lung cancer. Int J Radiat Oncol Biol Phys. 2006;65:726–32.PubMedCrossRef
6.
Zurück zum Zitat Bradley J, Thorstad WL, Mutic S, Miller TR, Dehdashti F, Siegel BA, et al. Impact of FDG-PET on radiation therapy volume delineation in non-small cell lung cancer. Int J Radiat Oncol Biol Phys. 2004;59:78–86.PubMedCrossRef Bradley J, Thorstad WL, Mutic S, Miller TR, Dehdashti F, Siegel BA, et al. Impact of FDG-PET on radiation therapy volume delineation in non-small cell lung cancer. Int J Radiat Oncol Biol Phys. 2004;59:78–86.PubMedCrossRef
7.
Zurück zum Zitat Hong R, Halama J, Bova D, Sethi A, Emami B. Correlation of PET standard uptake value and CT window-level thresholds for target volume delineation in CT-based radiation treatment planning. Int J Radiat Oncol Biol Phys. 2007;67:720–6.PubMedCrossRef Hong R, Halama J, Bova D, Sethi A, Emami B. Correlation of PET standard uptake value and CT window-level thresholds for target volume delineation in CT-based radiation treatment planning. Int J Radiat Oncol Biol Phys. 2007;67:720–6.PubMedCrossRef
8.
Zurück zum Zitat Paulino AC, Johnstone PA. FDG-PET in radiotherapy treatment planning: Pandora’s box? Int J Radiat Oncol Biol Phys. 2004;59:4–5.PubMedCrossRef Paulino AC, Johnstone PA. FDG-PET in radiotherapy treatment planning: Pandora’s box? Int J Radiat Oncol Biol Phys. 2004;59:4–5.PubMedCrossRef
9.
Zurück zum Zitat Brianzoni E, Rossi G, Ancidei S, Berbellini A, Capoccetti F, Cidda C, et al. Radiotherapy planning: PET/CT scanner performances in the definition of gross tumor volume and clinical target volume. Eur J Nucl Med Mol Imaging. 2005;32:1392–9.PubMedCrossRef Brianzoni E, Rossi G, Ancidei S, Berbellini A, Capoccetti F, Cidda C, et al. Radiotherapy planning: PET/CT scanner performances in the definition of gross tumor volume and clinical target volume. Eur J Nucl Med Mol Imaging. 2005;32:1392–9.PubMedCrossRef
10.
Zurück zum Zitat Ciernik IF, Dizendorf E, Baumert BG, Reiner B, Burger C, Davis JB, et al. Radiation treatment planning with an integrated positron emission and computed tomography (PET/CT): a feasibility study. Int J Radiat Oncol Biol Phys. 2003;57:853–63.PubMedCrossRef Ciernik IF, Dizendorf E, Baumert BG, Reiner B, Burger C, Davis JB, et al. Radiation treatment planning with an integrated positron emission and computed tomography (PET/CT): a feasibility study. Int J Radiat Oncol Biol Phys. 2003;57:853–63.PubMedCrossRef
11.
Zurück zum Zitat Mah K, Caldwell CB, Ung YC, Danjoux CE, Balogh JM, Ganguli SN, et al. The impact of (18)FDG-PET on target and critical organs in CT-based treatment planning of patients with poorly defined non-small-cell lung carcinoma: a prospective study. Int J Radiat Oncol Biol Phys. 2002;52:339–50.PubMedCrossRef Mah K, Caldwell CB, Ung YC, Danjoux CE, Balogh JM, Ganguli SN, et al. The impact of (18)FDG-PET on target and critical organs in CT-based treatment planning of patients with poorly defined non-small-cell lung carcinoma: a prospective study. Int J Radiat Oncol Biol Phys. 2002;52:339–50.PubMedCrossRef
12.
Zurück zum Zitat Miller TR, Grigsby PW. Measurement of tumor volume by PET to evaluate prognosis in patients with advanced cervical cancer treated by radiation treatment. Int J Radiat Oncol Biol Phys. 2002;53:353–9.PubMedCrossRef Miller TR, Grigsby PW. Measurement of tumor volume by PET to evaluate prognosis in patients with advanced cervical cancer treated by radiation treatment. Int J Radiat Oncol Biol Phys. 2002;53:353–9.PubMedCrossRef
13.
Zurück zum Zitat Paulino AC, Koshy M, Howell R, Schuster D, Davis LW. Comparison of CT- and FDG-PET-defined gross tumor volume in intensity-modulated radiotherapy for head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2005;61:1385–92.PubMedCrossRef Paulino AC, Koshy M, Howell R, Schuster D, Davis LW. Comparison of CT- and FDG-PET-defined gross tumor volume in intensity-modulated radiotherapy for head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2005;61:1385–92.PubMedCrossRef
14.
Zurück zum Zitat Daisne JF, Sibomana M, Bol A, Doumont T, Lonneux M, Gregoire V. Tri-dimensional automatic segmentation of PET volumes based on measured source-to-background ratios: influence of reconstruction algorithms. Radiother Oncol. 2003;69:247–50.PubMedCrossRef Daisne JF, Sibomana M, Bol A, Doumont T, Lonneux M, Gregoire V. Tri-dimensional automatic segmentation of PET volumes based on measured source-to-background ratios: influence of reconstruction algorithms. Radiother Oncol. 2003;69:247–50.PubMedCrossRef
15.
Zurück zum Zitat Schinagl DA, Vogel WV, Hoffmann AL, van Dalen JA, Oyen WJ, Kaanders JH. Comparison of five segmentation tools for 18F-fluoro-deoxy-glucose-positron emission tomography-based target volume definition in head and neck cancer. Int J Radiat Oncol Biol Phys. 2007;69:1282–9.PubMedCrossRef Schinagl DA, Vogel WV, Hoffmann AL, van Dalen JA, Oyen WJ, Kaanders JH. Comparison of five segmentation tools for 18F-fluoro-deoxy-glucose-positron emission tomography-based target volume definition in head and neck cancer. Int J Radiat Oncol Biol Phys. 2007;69:1282–9.PubMedCrossRef
16.
Zurück zum Zitat Vogel WV, Wensing BM, van Dalen JA, Krabbe PF, van den Hoogen FJ, Oyen W. Optimised PET reconstruction of the head and neck area: improved diagnostic accuracy. Eur J Nucl Med Mol Imaging. 2005;32:1276–82.PubMedCrossRef Vogel WV, Wensing BM, van Dalen JA, Krabbe PF, van den Hoogen FJ, Oyen W. Optimised PET reconstruction of the head and neck area: improved diagnostic accuracy. Eur J Nucl Med Mol Imaging. 2005;32:1276–82.PubMedCrossRef
17.
Zurück zum Zitat Vogel WV, Schinagl DA, van Dalen JA, Kaanders JH, Oyen WJ. Validated image fusion of dedicated PET and CT for external beam radiation therapy in the head and neck area. Q J Nucl Med Mol Imaging. 2008;52:74–83.PubMed Vogel WV, Schinagl DA, van Dalen JA, Kaanders JH, Oyen WJ. Validated image fusion of dedicated PET and CT for external beam radiation therapy in the head and neck area. Q J Nucl Med Mol Imaging. 2008;52:74–83.PubMed
18.
Zurück zum Zitat Schinagl DA, Hoffmann AL, Vogel WV, van Dalen JA, Verstappen SM, Oyen WJ, et al. Can FDG-PET assist in radiotherapy target volume definition of metastatic lymph nodes in head-and-neck cancer? Radiother Oncol. 2009;91:95–100.PubMedCrossRef Schinagl DA, Hoffmann AL, Vogel WV, van Dalen JA, Verstappen SM, Oyen WJ, et al. Can FDG-PET assist in radiotherapy target volume definition of metastatic lymph nodes in head-and-neck cancer? Radiother Oncol. 2009;91:95–100.PubMedCrossRef
19.
Zurück zum Zitat Brun E, Kjellén E, Tennvall J, Ohlsson T, Sandell A, Perfekt R, et al. FDG PET studies during treatment: prediction of therapy outcome in head and neck squamous cell carcinoma. Head Neck. 2002;24:127–35.PubMedCrossRef Brun E, Kjellén E, Tennvall J, Ohlsson T, Sandell A, Perfekt R, et al. FDG PET studies during treatment: prediction of therapy outcome in head and neck squamous cell carcinoma. Head Neck. 2002;24:127–35.PubMedCrossRef
20.
Zurück zum Zitat Halfpenny W, Hain SF, Biassoni L, Maisey MN, Sherman JA, McGurk M. FDG-PET. A possible prognostic factor in head and neck cancer. Br J Cancer. 2002;86:512–6.PubMedCrossRef Halfpenny W, Hain SF, Biassoni L, Maisey MN, Sherman JA, McGurk M. FDG-PET. A possible prognostic factor in head and neck cancer. Br J Cancer. 2002;86:512–6.PubMedCrossRef
21.
Zurück zum Zitat Lee SW, Nam SY, Im KC, Kim JS, Choi EK, Ahn SD, et al. Prediction of prognosis using standardized uptake value of 2-[(18)F]fluoro-2-deoxy-d-glucose positron emission tomography for nasopharyngeal carcinomas. Radiother Oncol. 2008;97:211–6.CrossRef Lee SW, Nam SY, Im KC, Kim JS, Choi EK, Ahn SD, et al. Prediction of prognosis using standardized uptake value of 2-[(18)F]fluoro-2-deoxy-d-glucose positron emission tomography for nasopharyngeal carcinomas. Radiother Oncol. 2008;97:211–6.CrossRef
22.
Zurück zum Zitat Machtay M, Natwa M, Andrel J, Hyslop T, Anne PR, Lavarino J, et al. Pretreatment FDG-PET standardized uptake value as a prognostic factor for outcome in head and neck cancer. Head Neck. 2009;31:195–201.PubMedCrossRef Machtay M, Natwa M, Andrel J, Hyslop T, Anne PR, Lavarino J, et al. Pretreatment FDG-PET standardized uptake value as a prognostic factor for outcome in head and neck cancer. Head Neck. 2009;31:195–201.PubMedCrossRef
23.
Zurück zum Zitat Minn H, Lapela M, Klemi PJ, Grénman R, Leskinen S, Lindholm P, et al. Prediction of survival with fluorine-18-fluoro-deoxyglucose and PET in head and neck cancer. J Nucl Med. 1997;38:1907–11.PubMed Minn H, Lapela M, Klemi PJ, Grénman R, Leskinen S, Lindholm P, et al. Prediction of survival with fluorine-18-fluoro-deoxyglucose and PET in head and neck cancer. J Nucl Med. 1997;38:1907–11.PubMed
24.
Zurück zum Zitat Roh JL, Pae KH, Choi SH, Kim JS, Lee S, Kim SB, et al. 2-[18F]-Fluoro-2-deoxy-D-glucose positron emission tomography as guidance for primary treatment in patients with advanced-stage resectable squamous cell carcinoma of the larynx and hypopharynx. Eur J Surg Oncol. 2007;33:790–5.PubMed Roh JL, Pae KH, Choi SH, Kim JS, Lee S, Kim SB, et al. 2-[18F]-Fluoro-2-deoxy-D-glucose positron emission tomography as guidance for primary treatment in patients with advanced-stage resectable squamous cell carcinoma of the larynx and hypopharynx. Eur J Surg Oncol. 2007;33:790–5.PubMed
25.
Zurück zum Zitat Schwartz DL, Rajendran J, Yueh B, Coltrera MD, Leblanc M, Eary J, et al. FDG-PET prediction of head and neck squamous cell cancer outcomes. Arch Otolaryngol Head Neck Surg. 2004;130:1361–7.PubMedCrossRef Schwartz DL, Rajendran J, Yueh B, Coltrera MD, Leblanc M, Eary J, et al. FDG-PET prediction of head and neck squamous cell cancer outcomes. Arch Otolaryngol Head Neck Surg. 2004;130:1361–7.PubMedCrossRef
26.
Zurück zum Zitat Chung MK, Jeong HS, Park SG, Jang JY, Son YI, Choi JY, et al. Metabolic tumor volume of [18F]-fluorodeoxyglucose positron emission tomography/computed tomography predicts short-term outcome to radiotherapy with or without chemotherapy in pharyngeal cancer. Clin Cancer Res. 2009;15:5861–8.PubMedCrossRef Chung MK, Jeong HS, Park SG, Jang JY, Son YI, Choi JY, et al. Metabolic tumor volume of [18F]-fluorodeoxyglucose positron emission tomography/computed tomography predicts short-term outcome to radiotherapy with or without chemotherapy in pharyngeal cancer. Clin Cancer Res. 2009;15:5861–8.PubMedCrossRef
27.
Zurück zum Zitat La TH, Filion EJ, Turnbull BB, Chu JN, Lee P, Nguyen K, et al. Metabolic tumor volume predicts for recurrence and death in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2009;74:1335–41.PubMedCrossRef La TH, Filion EJ, Turnbull BB, Chu JN, Lee P, Nguyen K, et al. Metabolic tumor volume predicts for recurrence and death in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2009;74:1335–41.PubMedCrossRef
28.
Zurück zum Zitat Seol YM, Kwon BR, Song MK, Choi YJ, Shin HJ, Chung JS, et al. Measurement of tumor volume by PET to evaluate prognosis in patients with head and neck cancer treated by chemo-radiation therapy. Acta Oncol. 2010;49:201–8.PubMedCrossRef Seol YM, Kwon BR, Song MK, Choi YJ, Shin HJ, Chung JS, et al. Measurement of tumor volume by PET to evaluate prognosis in patients with head and neck cancer treated by chemo-radiation therapy. Acta Oncol. 2010;49:201–8.PubMedCrossRef
29.
Zurück zum Zitat Soto DE, Kessler ML, Piert M, Eisbruch A. Correlation between pretreatment FDG-PET biological target volume and anatomical location of failure after radiation therapy for head and neck cancers. Radiother Oncol. 2008;89:13–8.PubMedCrossRef Soto DE, Kessler ML, Piert M, Eisbruch A. Correlation between pretreatment FDG-PET biological target volume and anatomical location of failure after radiation therapy for head and neck cancers. Radiother Oncol. 2008;89:13–8.PubMedCrossRef
30.
Zurück zum Zitat Suzuki K, Nishioka T, Homma A, Tsuchiya K, Yasuda M, Aoyama H, et al. Value of fluorodeoxyglucose positron emission tomography before radiotherapy for head and neck cancer: does standardized value predict treatment outcome? Jpn J Radiol. 2009;27:237–42.PubMedCrossRef Suzuki K, Nishioka T, Homma A, Tsuchiya K, Yasuda M, Aoyama H, et al. Value of fluorodeoxyglucose positron emission tomography before radiotherapy for head and neck cancer: does standardized value predict treatment outcome? Jpn J Radiol. 2009;27:237–42.PubMedCrossRef
31.
Zurück zum Zitat Thorwarth D, Eschmann SM, Holzner F, Paulsen F, Alber M. Combined uptake of [18F]FDG and [18F]FMISO correlates with radiation treatment outcome in head and neck cancer patients. Radiother Oncol. 2006;80:151–6.PubMedCrossRef Thorwarth D, Eschmann SM, Holzner F, Paulsen F, Alber M. Combined uptake of [18F]FDG and [18F]FMISO correlates with radiation treatment outcome in head and neck cancer patients. Radiother Oncol. 2006;80:151–6.PubMedCrossRef
32.
Zurück zum Zitat Vernon MR, Maheshwari M, Schultz CJ, Michel MA, Wong SJ, Campbell BH, et al. Clinical outcomes of patients receiving integrated PET/CT-guided radiotherapy for head and neck carcinoma. Int J Radiat Oncol Biol Phys. 2008;70:678–84.PubMedCrossRef Vernon MR, Maheshwari M, Schultz CJ, Michel MA, Wong SJ, Campbell BH, et al. Clinical outcomes of patients receiving integrated PET/CT-guided radiotherapy for head and neck carcinoma. Int J Radiat Oncol Biol Phys. 2008;70:678–84.PubMedCrossRef
33.
Zurück zum Zitat Hermans R, Op de Beeck K, Van den Bogaert W, Rijnders A, Staelens L, Feron M, et al. The relation of CT-determined tumor parameters and local and regional outcome of tonsillar cancer after definitive radiation treatment. Int J Radiat Oncol Biol Phys. 2001;50:37–45.PubMedCrossRef Hermans R, Op de Beeck K, Van den Bogaert W, Rijnders A, Staelens L, Feron M, et al. The relation of CT-determined tumor parameters and local and regional outcome of tonsillar cancer after definitive radiation treatment. Int J Radiat Oncol Biol Phys. 2001;50:37–45.PubMedCrossRef
34.
Zurück zum Zitat Nathu RM, Mancuso AA, Zhu TC, Mendenhall WM. The impact of primary tumor volume on local control for oropharyngeal squamous cell carcinoma treated with radiotherapy. Head Neck. 2000;22:1–5.PubMedCrossRef Nathu RM, Mancuso AA, Zhu TC, Mendenhall WM. The impact of primary tumor volume on local control for oropharyngeal squamous cell carcinoma treated with radiotherapy. Head Neck. 2000;22:1–5.PubMedCrossRef
35.
Zurück zum Zitat Chao KS, Ozyigit G, Blanco AI, Thorstad WL, Deasy JO, Haughey BH, et al. Intensity-modulated radiation therapy for oropharyngeal carcinoma: impact of tumor volume. Int J Radiat Oncol Biol Phys. 2004;59:43–50.PubMedCrossRef Chao KS, Ozyigit G, Blanco AI, Thorstad WL, Deasy JO, Haughey BH, et al. Intensity-modulated radiation therapy for oropharyngeal carcinoma: impact of tumor volume. Int J Radiat Oncol Biol Phys. 2004;59:43–50.PubMedCrossRef
36.
Zurück zum Zitat Hoogsteen IJ, Marres HA, Wijffels KI, Rijken PF, Peters JP, van den Hoogen FJ, et al. Colocalization of carbonic anhydrase 9 expression and cell proliferation in human head and neck squamous cell carcinoma. Clin Cancer Res. 2005;11:97–106.PubMed Hoogsteen IJ, Marres HA, Wijffels KI, Rijken PF, Peters JP, van den Hoogen FJ, et al. Colocalization of carbonic anhydrase 9 expression and cell proliferation in human head and neck squamous cell carcinoma. Clin Cancer Res. 2005;11:97–106.PubMed
37.
Zurück zum Zitat Hermans R. Head and neck cancer: how imaging predicts treatment outcome. Cancer Imaging. 2006;6:S145–53.PubMedCrossRef Hermans R. Head and neck cancer: how imaging predicts treatment outcome. Cancer Imaging. 2006;6:S145–53.PubMedCrossRef
38.
Zurück zum Zitat Troost EG, Schinagl DA, Bussink J, Boerman OC, van der Kogel AJ, Oyen WJ, et al. Innovations in radiotherapy planning of head and neck cancers: role of PET. J Nucl Med. 2010;51:66–76.PubMedCrossRef Troost EG, Schinagl DA, Bussink J, Boerman OC, van der Kogel AJ, Oyen WJ, et al. Innovations in radiotherapy planning of head and neck cancers: role of PET. J Nucl Med. 2010;51:66–76.PubMedCrossRef
Metadaten
Titel
Can FDG PET predict radiation treatment outcome in head and neck cancer? Results of a prospective study
verfasst von
Dominic A. X. Schinagl
Paul N. Span
Wim J. Oyen
Johannes H. A. M. Kaanders
Publikationsdatum
01.08.2011
Verlag
Springer-Verlag
Erschienen in
European Journal of Nuclear Medicine and Molecular Imaging / Ausgabe 8/2011
Print ISSN: 1619-7070
Elektronische ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-011-1789-x

Weitere Artikel der Ausgabe 8/2011

European Journal of Nuclear Medicine and Molecular Imaging 8/2011 Zur Ausgabe