Skip to main content
Erschienen in: Annals of Nuclear Medicine 4/2017

Open Access 15.03.2017 | Original Article

Serial imaging using [18F]Fluorodeoxyglucose positron emission tomography and histopathologic assessment in predicting survival in a population of surgically resectable distal oesophageal and gastric adenocarcinoma following neoadjuvant therapy

Erschienen in: Annals of Nuclear Medicine | Ausgabe 4/2017

Abstract

Background and objectives

We retrospectively evaluated the value of PET/CT in predicting survival and histopathological tumour-response in patients with distal oesophageal and gastric adenocarcinoma following neoadjuvant treatment.

Methods

Twenty-one patients with resectable distal oesophageal adenocarcinoma and 14 with gastric adenocarcinoma between January 2002 and December 2011, who had undergone serial PET before and after neoadjuvant therapy followed by surgery, were enrolled. Maximum standard uptake value (SUVmax) and metabolic tumour volume were measured and correlated with tumour regression grade and survival.

Results

Histopathological tumour response (PR) is a stronger predictor of overall and disease-free survival compared to metabolic response. ∆%SUVmax ≥70% was the only PET metric that predicted PR (82.4% sensitivity, 61.5% specificity, p = 0.047). Histopathological non-responders had a higher risk of death (HR 8.461, p = 0.001) and recurrence (HR 6.385, p = 0.002) and similarly in metabolic non-responders for death (HR 2.956, p = 0.063) and recurrence (HR 3.614, p = 0.028). Ordinalised ∆%SUVmax showed a predictive trend for OS and DFS, but failed to achieve statistical significance.

Conclusions

PR was a stronger predictor of survival than metabolic response. ∆%SUVmax ≥70% was the best biomarker on PET that predicted PR and survival in oesophageal and gastric adenocarcinoma. Ordinalisation of ∆%SUVmax was not helpful in predicting primary outcomes.

Introduction

Despite the increase in the incidence of oesophageal adenocarcinoma since 1988 and the steady decline in the incidence of gastric adenocarcinoma since 1982, oesophageal and gastric malignancies collectively accounted for 2317 deaths in Australia in 2010 [1]. While surgery remains the mainstay of curative treatment, the administration of neoadjuvant multimodality therapy has been shown to increase rates of histopathological complete response and entail modest survival benefit over surgery alone [2]. Positron emission tomography (PET) using 18F-2-fluoro-2-deoxyglucose (18F-FDG) permits in vivo characterisation of pathological 18F-FDG retention and PET has gained acceptance for initial staging of oesophageal malignancy by improving the detection of occult distant metastases [3]. Recently, several authors have investigated the predictive value of PET metrics for early response to therapy in oesophageal and gastric neoplasms, given that PET has been shown to be useful in other cancers [47].
Tumour regression grades (TRG), a measure of histopathological tumour response (PR) based on an estimation of the percentage of residual tumour tissue in relation to the macroscopically identifiable tumour bed at the primary site, was adopted from studies conducted on gastric cancers [8] and oesophageal adenocarcinomas [9]. Lordick, et al. [10] validated the use of therapy-induced changes in PET metrics to predict PR and to stratify distal oesophageal and gastric cardia adenocarcinoma patients into different prognostic groups. They concluded that 35% regression of maximum standardised uptake value (SUVmax), a semi-quantitative measure of 18F-FDG retention in the primary tumour bed was the optimal cut-off to identify histopathological responders with 100% sensitivity and 58% specificity. However, the evidence regarding the predictive value of PET-based biomarkers in gastric cancers is limited and inconsistent. One prospective study on advanced gastric cancer patients confirmed the highly predictive value of 35% reduction in SUVmax on PR with 77% sensitivity and 86% specificity [11], while one small study comprising locally advanced gastric cancers showed a 45% decrease in SUV from baseline and day 35 significantly predicted PR whereas the change at day 15 did not [12].
There is increasing interest in investigating the prognostic value of PET volumetric parameters such as metabolic tumour volume (MTV), but the utility of this more novel imaging biomarker is experimental. One study reported that a 63% reduction in MTV was the optimal cut-off to identify histopathologically responding distal oesophageal adenocarcinoma with 91% sensitivity and 90% specificity [13]. The evidence on the predictive value of MTV in gastric cancer is scarce.
Several authors have validated that metabolic responders (MR+) identified on PET had better prognosis [10, 11, 14]. Some studies have shown variable thresholds [15] in predicting PR and survival on PET, while others failed to validate the predictive power of PET metrics [16, 17]. Lee did not find a correlation between the change in SUVmax of the primary tumour with PR after neoadjuvant chemotherapy and surgery in gastric carcinoma [18]. Vallbohmer also did not find a correlation between a change in SUVmax (baseline and post-treatment PET at 2 weeks) and PR (<10% viable cells) or overall survival in oesophageal carcinoma [19]. This study aims to retrospectively evaluate the performance of PET in predicting survival and PR to neoadjuvant therapy in patients with surgically resected distal oesophageal and gastric adenocarcinoma.

Method

Patient population

Fifty-six patients with newly diagnosed resectable distal oesophageal and gastric adenocarcinoma who underwent neoadjuvant therapy and serial 18F-FDG PET scans at Liverpool Hospital (Sydney, Australia) between January 2002 and December 2011 were included. 35 patients (21 patients with distal oesophageal adenocarcinoma and 14 patients with gastric adenocarcinoma) with sufficient PET data and TRG scores were analysed (Fig. 1). Patients with non-resectable disease at diagnosis, non-FDG-avid primary tumour on initial PET scan (PET-1), insufficient PET data, active concurrent cancer unrelated to oesophageal and gastric cancer, previous neoadjuvant therapy prior to initial PET and those who did not undergo surgery were excluded. Post-treatment PET (PET-2) parameters were not evaluated in one patient with a significant difference in uptake time between the two PET scans (64 min) and in five patients who had oesophageal stents implanted after PET-1. Most patients underwent PET/CT scans apart from two patients where PET-alone imaging was performed.
Tumour staging was based on the sixth edition of the American Joint Committee of Cancer (AJCC) Staging Manual [20], but classification of tumour location was based on the seventh edition [21].

Patient information

Demographic, clinical and follow-up data were gathered through various databases including the South Western Sydney Area Health Service’s (SWSAHS) online patient information system, Cancer Therapy Centre (MOSAIQ), Departments of Surgery and Nuclear Medicine and PET.

PET imaging

All patients underwent a baseline PET scan for staging (PET-1) and a post-treatment PET scan (PET-2). PET-alone scans operating in three-dimensional mode (Allegro, Philips Medical Systems, Milpitas, CA, US) with germanium source attenuation were performed prior to February 2006, and PET/CT scans (Gemini GXL-6, Philips Medical Systems, Miltipas, CA, US) using low-dose CT without contrast enhancement for attenuation correction.
A standardised protocol comprised a minimum 4-h fasting period and blood glucose levels <10 mmol/L prior to 18F-FDG (5.14 MBq/kg), administration intravenously. Patients were scanned after an uptake period of approximately 60 min.

PET data analysis

PET scans were analysed by two accredited Nuclear Medicine physicians in consensus (M.L. and J.Y.) according to a standardised protocol where the SUVmax was measured using a 15 mm wide region of interest around the primary tumour. MTV was measured using vendor’s software with a SUVmax threshold that best delineated the tumour. Scans (PET-1 and PET-2) were analysed blinded from all clinical, pathological and imaging data apart from the knowledge that all patients had oesophageal and gastric malignancy and had completed neoadjuvant therapy. PET-1 and PET-2 measurements and any absolute and relative differences in 18F-FDG uptake were correlated with TRG and survival. If no residual tumour was visible and uptake was indistinguishable from background oesophageal or gastric activity on post-treatment scan, no volumetric measurement was attempted and the percentage reduction in abnormal tracer uptake is assigned 100%. Metabolic responders (MR+) are patients with ∆%SUVmax ≥70%.

Histopathological response evaluation

Surgical specimens were retrospectively examined by a single pathologist (S.L.). TRG score was assessed semi-quantitatively into either complete (TRG 1a: no residual tumour), subtotal (TRG 1b: <10% of residual tumour), partial (TRG 2: 10–50% of residual tumour) and minimal response (TRG 3: >50% residual tumour) based on Becker et al. [8]. The pathologist was blinded from all clinical, pathological and imaging data. Patients with complete or subtotal tumour regression were classified as histopathological responders (PR+). All other patients were classified as non-responders (PR).

Follow-up

Disease-status and survival status at the time of census were recorded. Overall survival (OS) was calculated from the date of PET-1 to date of death or date of most recent follow-up. Disease-free survival (DFS) was calculated from date of surgery to the date of confirmed recurrence. If death was a direct consequence of surgery within 2 weeks of surgery, then the patient was excluded from survival analyses.

Statistical analysis

Absolute numbers and percentages were computed to describe the patient population, and quantitative values are expressed as median and range. Chi-square test was used to examine associations between categorical variables. Receiver operator characteristics (ROC) curve was performed to find the optimal cut-offs of the PET parameters. Survival curves were generated using Kaplan–Meier estimates and significance of difference between curves was tested with log-rank tests. Univariate analysis of survival was performed using Cox regression analysis and the estimated hazard ratio (HR) and 95% confidence interval (CI) were reported. All statistical analyses were performed using IBM SPSS Statistics 21 and p < 0.05 was considered statistically significant.

Results

Patient characteristics

There was a preponderance of male subjects and neoadjuvant chemotherapy-alone treatment in the cohort (Table 1). Most patients were node negative on staging PET.
Table 1
Patient characteristics and clinicopathological parameters
Parameters
n (%)
Gender
 Males
30 (85.7)
 Females
5 (14.3)
Median age at diagnosis
61.7 years (40.6–74.5)
Histology
 Signet ring cell
4 (11.4)
 Non-Signet ring cell
31 (88.6)
Tumour Location (AJCC 7th ed.)
 Distal oesophagus
18 (51.4)
 Stomach
17 (48.6)
  Cardia of stomach
9
  Fundus
1
  Body of stomach
4
  Antrum
3
  Pylorus
0
Tumour Grade
 Moderate
17 (48.6)
 Poor
18 (51.4)
Clinical T stage at diagnosis (AJCC 6th ed.)
 T2
3 (8.6)
 T3
23 (65.7)
 T4
6 (17.1)
 Missing
3 (8.6)
Nodal involvement on staging PET
 N
26 (74.3)
 N+
9 (25.7)
Overall clinical stage at diagnosis (AJCC 6th ed.)
 I
2 (5.7)
 II
12 (48.6)
 III
20 (55.1)
 Missing
1 (2.9)
Type of neoadjuvant therapy
 CT alone
20 (57.1)
 CRT
9 (25.7)
 RT alone
3 (8.6)
 Not specified
3 (8.6)
TRG
 1a
3 (8.6)
 1b
13 (37.1)
 2
4 (11.4)
 3
15 (42.9)
Recurrence
 No
18 (51.4)
 Yes
17 (48.6)
Median disease-free survival
13.8 months (2.1-122.4)
Dead
 No
15 (42.9)
 Yes
20 (57.1)
Median overall survival
22.3 months (9.6-122.4)
Median follow-up period
22.7 months (10.8-122.4)
Two-year overall survival rate
40.0%
T stage, depth of invasion, N stage nodal involvement, PET positron emission tomography, AJCC American Joint Committee of Cancer Staging Manual, N negative nodal involvement, N + positive nodal involvement; CT chemotherapy, CRT chemoradiotherapy, RT radiotherapy, TRG tumour regression grade
There was a significant difference (p = 0.002) in the median patient weight at PET-1 (79 kg, 48–115) compared to PET-2 of (76 kg, 55–118). Median uptake time at PET-1 was 63 min and for PET-2 was 64 min (p = 0.878) with a median difference in uptake times of 6 min (0–22). The median interval between the two PET scans was 106 days (55–153).

PET and histopathological tumour response evaluation

The relative reduction in SUVmax (∆%SUVmax) ≥70% was the only optimum cut-off to predict PR (p = 0.047) on ROC analysis with 82.4% sensitivity and 61.5% specificity. There was a trend for a greater proportion of PR+ having an absolute reduction in SUVmax (∆SUVmax) ≥5.75 (p = 0.071). Other PET metrics did not retain statistical significance and were dichotomised at the respective median values (Table 2). By ordinalising ∆%SUVmax into a schema similar to that of TRG with complete response (∆%SUVmax = 100%), subtotal response (∆%SUVmax ≥70%), partial response (∆%SUVmax ≥35%) and minimal response (∆%SUVmax <35%), no significant result was attained.
Table 2
Predictive value of various PET parameters of tumour regression grade (TRG) score
 
n (%)
Chi-square
p
TRG1a–1b
TRG2–3
Baseline SUVmax
 SUV1max ≥9.70
10 (28.6)
10 (28.6)
0.345
0.557
 SUV1max <9.70
6 (17.1)
9 (25.7)
  
Post-treatment SUVmax
 SUV2max ≥3.75
5 (17.2)
8 (27.6)
0.386
0.534
 SUV2max <3.75
8 (27.6)
8 (27.6)
  
Metabolic response based on ∆SUVmax
 ∆SUVmax ≥5.75
10 (34.5)
7 (24.2)
3.254
0.071
 ∆SUVmax <5.75
3 (10.3)
9 (31.0)
  
Metabolic response based on ROC analysis of ∆%SUVmax
 ∆%SUVmax ≥70%
8 (27.6)
4 (13.8)
3.948
0.047
 ∆%SUVmax <70%
5 (17.2)
12 (41.4)
  
Baseline MTV (cm3)
 MTV1 ≥47.30
8 (27.6)
10 (34.5)
0.024
1.000
 MTV1 <47.30
8 (27.6)
9 (31.0)
  
Post-treatment MTV (cm3)
 MTV2 ≥12.00
3 (11.0)
7 (22.8)
1.008
0.315
 MTV2 <12.00
8 (27.6)
8 (27.6)
  
 Missing
3 (11.0)
   
Metabolic response based on ∆MTV
 ∆MTV ≥39.40
7 (22.8)
9 (31.0)
0.035
0.851
 ∆MTV <39.40
4 (13.2)
6 (22.0)
  
 Missing
3 (11.0)
   
Metabolic response based on ∆% MTV
 ∆%MTV ≥80%
5 (14.0)
6 (22.0)
0.077
0.781
 ∆%MTV <80%
6 (22.0)
9 (31.0)
  
 Missing
3 (11.0)
   
Metabolic response based on various cut-offs of ∆%SUVmax
 ∆%SUVmax = 100%
2 (6.9)
2 (6.9)
3.883
0.274
 ∆%SUVmax ≥70–99%
6 (20.7)
2 (6.9)
 
 ∆%SUVmax ≥35–69%
5 (17.2)
9 (31.1)
 
 ∆%SUVmax <35%
0 (0.0)
3 (10.3)
 
Statistical significant result is in bold
Post-treatment scans in 5 patients were excluded from analysis due to oesophageal stent insertion. One patient was excluded due to significant difference in uptake times between the two scans
PET positron emission tomography, TRG tumour regression grade, SUV1 max baseline SUVmax, SUV2 max post-treatment SUVmax, ∆SUV max absolute reduction in SUVmax, ∆%SUV max relative reduction in SUVmax, MTV metabolic tumour volume, MTV1 baseline MTV, MTV2 post-treatment MTV, ∆MTV absolute reduction in MTV, ∆%MTV relative reduction in MTV

PET, TRG and survival analysis

MR+ and PR+ had a significantly longer OS and DFS than their non-responding counterparts (Fig. 2). Median OS and DFS were not reached in MR+.
PR had a significantly greater risk of death (HR 8.461; p = 0.001) and recurrence (HR 6.385; p = 0.002) (Table 3). Similarly, metabolic non-responders (MR) with ∆%SUVmax <70% entailed a significantly greater risk of death (HR 2.956, p = 0.063) and recurrence (HR 3.614; p = 0.028) (Table 3). PR was a stronger predictor of OS (HR 8.461 vs 2.956) and DFS (HR 6.385 vs 3.614) compared to ∆%SUVmax≥70%.
Table 3
Prognostic value of metabolic parameters on PET and histopathological tumour response for survival
 
n (%)
Univariate Cox regression test
Univariate Cox regression test
OS HR
95% CI
p
DFS HR
95% CI
p
Baseline SUVmax
 SUV1max ≥9.70
20 (57.1)
1.000
 
0.044
1.000
 
0.030
 SUV1max < 9.70
15 (42.9)
2.589
1.025–6.535
 
2.797
1.107–7.071
 
Post-treatment SUVmax
 SUV2max ≥3.75
13 (44.8)
1.000
 
0.376
1.000
 
0.352
 SUV2max < 3.75
16 (55.2)
1.627
0.554–4.780
 
1.658
0.572–4.812
 
Metabolic response based on ∆SUVmax
 ∆SUVmax ≥ 5.75
17 (58.6)
1.000
 
0.112
1.000
 
0.066
 ∆SUVmax < 5.75
12 (41.2)
2.223
0.829–6.013
 
2.563
0.941–6.981
 
Metabolic response based on ROC analysis of ∆%SUVmax
 ∆%SUVmax ≥70%
12 (41.2)
1.000
 
0.063
1.000
 
0.028
 ∆%SUVmax <70%
17 (58.6)
2.956
0.945–9.247
 
3.614
1.150–11.533
 
Baseline MTV (cm3)
 MTV1 ≥47.30
18 (51.4)
1.000
 
0.141
1.000
 
0.133
 MTV1 < 47.30
17 (48.6)
1.996
0.795–5.013
 
2.031
0.806–5.118
 
Post-treatment MTV (cm3)
 MTV2 ≥12.00
10 (34.5)
1.000
 
0.817
1.000
 
0.795
 MTV2 < 12.00
16 (55.2)
0.860
0.241–3.074
 
0.845
0.238–3.003
 
 Missing
3 (10.3)
      
Metabolic response based on ∆MTV
 ∆MTV ≥39.40
16 (55.2)
1.000
 
0.305
1.000
 
0. 362
 ∆MTV < 39.40
10 (34.5)
1.182
0.583–5.639
 
1.698
0.544–5.296
 
 Missing
3 (10.3)
      
Metabolic response based on ∆% MTV
 ∆%MTV ≥80%
10 (34.5)
1.000
 
0.707
1.000
 
0. 681
 ∆%MTV < 80%
16 (55.2)
0.792
0.235–2.672
 
0.775
0.231–2.605
 
 Missing
3 (10.3)
      
Metabolic response based on various cut-offs of ∆%SUVmax
 ∆%SUVmax = 100%
4 (13.8)
1.000
  
1.000
  
 ∆%SUVmax ≥70–99%
8 (27.6)
0.402
0.056–2.898
0.366
0.454
0.063–3.247
0.431
 ∆%SUVmax ≥35–69%
14 (48.3)
1.552
0.331–7.727
0.577
2.137
0.460–9.922
0.332
 ∆%SUVmax <35%
3 (10.3)
3.241
0.429–24.495
0.254
3.718
0.491–28.183
0.204
Histopathological tumour response
 TRG1a–1b
16 (45.7)
1.000
  
1.000
  
 TRG2–3
19 (54.3)
8.461
2.355–30.396
0.001
6.385
2.019–20.195
0.002
Statistical significant result is in bold
Post-treatment scans in five patients were excluded from analysis due to oesophageal stent insertion. One patient was excluded due to significant difference in uptake times between the two scans
PET positron emission tomography, OS overall survival, DFS Disease-free survival, HR Hazard ratio, CI confidence interval, n number of patients, SUV1 max baseline SUVmax, SUV2 max post-treatment SUVmax, ∆SUV max absolute reduction in SUVmax, ∆%SUV max relative reduction in SUVmax, MTV metabolic tumour volume, MTV1 Baseline MTV, MTV2 post-treatment MTV, ∆MTV absolute reduction in MTV, ∆%MTV relative reduction in MTV, TRG tumour regression grade
For PET-1, baseline SUVmax <9.70 was the only statistically significant predictor of poor OS (HR 2.589, p = 0.044). The ∆%SUVmax <70% cut-off showed a trend (p = 0.063) towards poorer OS. Both groups reported a higher risk of recurrence (p = 0.030; p = 0.028) (Table 3). All other PET metrics were not statistically significant.
When we analysed TRG as a binary parameter-complete or subtotal (TRG 1a and TRG 1b), and partial or minimal response (TRG 2 and 3), TRG was significantly associated with survival status (p = 0.002) on Chi-square analysis.

Discussion

The correlations between TRG, survival and PET-metrics have not been widely reported in oesophageal and gastric adenocarcinomas. This study comprised a heterogeneous group with resectable adenocarcinoma of the distal oesophagus and stomach. Although surgical treatment of oesophageal and gastric cancers occurs in relatively low volume centres in Australia, we have equivalent short and long term outcomes compared to Asian and European institutions [22, 23]. Therefore, our findings on the predictive performance of PET for survival and PR in this group are applicable to other populations. The study is also strengthened with centralized PET scan interpretation in consensus.
We found that a favourable metabolic response is associated with favourable PR. A reduction of SUVmax of the primary ≥70% following neoadjuvant treatment was associated with a TRG1a–1b. Our study showed that ∆%SUVmax ≥70% (p = 0.047) was the optimal cut-off that characterised PR with 82.4% sensitivity and 61.5% specificity. To our knowledge, the association between PR and PET metrics have not been studied extensively in gastric cancers. This is in line with a small study of 35 patients using PET-alone system which demonstrated ∆%SUVmax ≥35% predicted survival in locally advanced gastric cancer [11]. Our data included only two patients who underwent PET-alone imaging and while we acknowledge PET-alone and PET-CT can produce differences in SUV due to different methods of attenuation correction, the difference is reported to be relatively small in the clinical setting for non-osseous lesions of a sufficient size (our cohort comprised mostly T3 or T4 tumours) [24, 25].
Although there was a trend for an absolute therapy-induced change in SUVmax (p = 0.07) in predicting PR, we showed relative reduction as a stronger predictor. This may be due to the variable physiologic background FDG uptake in the oesophagus and stomach and relative reduction appears to be a better metric to gauge response. There is currently no consensus on the optimal thresholds in determining metabolic response on PET and various investigators have found different cut-offs [10, 17, 26, 27]. This most likely reflects different camera specifications and methodology and a standardised protocol in future prospective trials is mandatory [28].
There are conflicting results regarding the use of PET metrics in predicting survival in gastric cancer [2931]. Our results demonstrated both MR+ and PR+ had significantly longer OS and DFS than non-responders (Fig. 2). Becker et al. [8] showed that PR+ (TRG 1a/1b) in 480 gastric adenocarcinoma patients had a significantly longer OS (128.6 months) compared to partial (61.9 months) and minimal (40.1 months) responders and similar conclusions were drawn from studies on distal oesophageal adenocarcinoma [9]. In our study, MR+ had a significantly longer OS (median NR vs 23.5 months) and longer DFS (median NR vs 16.5 months). These findings are similar to those from the MUNICON I trial [10]. There was a trend for MR to have an almost threefold increase in risk of death (p = 0.06) and a significantly shorter DFS (p = 0.028) (Table 3). Although high SUVmax in many tumours have been shown to be poor prognosticators [32], in our study a high baseline SUVmax (>9.7) did not predict poor OS. This observation has also been shown in one study where patients with metastatic disease had lower SUVmax and SUVmean compared to M0 patients [33] and in another, no significant difference was found between limited and disseminated gastric cancers [34]. This may be due to several poor prognostic histological sub-types having low glucose metabolism, e.g. signet-ring cell or mucinous adenocarcinoma. Recent evidence also suggests a relationship between FDG avidity and HER2 expression and PET may have the potential to predict tumour phenotype [35].
PR+ in our study had a greater reduction in tumour 18F-FDG metabolism, highlighting the potential to formulate PET-guided treatment algorithm, which has been validated in the MUNICON II trial [36]. The greater histopathological remission rate among MR of the MUNICON II trial compared to those in the MUNICON I trial was attributed to the PET-based early metabolic assessment and subsequent escalation of therapy in MR from chemotherapy-alone to chemoradiotherapy [10, 36]. MR in the MUNICON I trial had their chemotherapy stopped after 2 weeks and went directly to surgical resection potentially avoiding toxicity from futile chemotherapy. Despite the addition of radiotherapy to cisplatin or 5-fluorouracil based chemotherapy in MUNICON II trial, MR still had a poor prognosis. A recent trial comprising a small number of patients suggested that changing chemotherapy regimens (to taxane-based) in PET non-responding patients may improve outcomes [37].
To our knowledge, this is the first study to ordinalise ∆%SUVmax similar to TRG scores using ∆%SUVmax ≥70% and ∆%SUVmax ≥35% cut-offs. Although it failed to achieve statistical significance, our analysis showed a predictive trend for DFS and OS (Table 3). This warrants further investigation in a larger cohort.
Volumetric measurements on PET is emerging as an important novel imaging biomarker in predicting prognosis in non-small cell lung (NSCLC) [7] and oesophageal cancers [13, 38], but there is limited evidence in gastric malignancies. In our study, neither the median MTV nor the MTV determined by ROC analysis (data not shown) predicted PR and survival, perhaps due to the difficulty and inaccuracy in delineating MTV using different SUV thresholds. This may partly be due to the variable physiological 18F-FDG uptake in gastric mucosa which attenuates the tumour to background ratio. Hence, MTV may not have the same prognostic value in gastric cancer compared with NSCLC or oesophageal cancer.
This retrospective study comprised only FDG-avid gastric tumours. It is well known that a significant proportion of gastric cancers can be falsely negative on FDG PET in particular in tumours rich in mucin and our data is not applicable to all gastric cancers [3941]. We combined patients with distal oesophageal and gastric cancers due to small sample size and a larger population could have allowed subgroup analyses.
In conclusion, PR was a stronger prognostic indicator than metabolic response, and ∆%SUVmax was the best PET-based biomarker that predicted PR and survival in oesophageal and gastric adenocarcinoma. This study highlighted the potential role of PET in optimising treatment protocols and allows non-responders to be detected early to have escalation of treatment in this poor prognostic group.

Compliance with ethical standards

Conflict of interest

The authors declare that there are no conflict of interests.

Ethical standard

Ethics approval was obtained from the Research and Ethics Office of the South Western Sydney Local Health District.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

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 Australian Institute of Health and Welfare [AIHW]. Cancer in Australia: an overview 2012. Canberra: Australian Institute of Health and Welfare, 2010. Australian Institute of Health and Welfare [AIHW]. Cancer in Australia: an overview 2012. Canberra: Australian Institute of Health and Welfare, 2010.
2.
Zurück zum Zitat Merett ND. Multimodality treatment of potentially curative gastric cancer: Geographical variations and future prospects. World J Gastroenterol. 2014;20(36):12892–9.CrossRef Merett ND. Multimodality treatment of potentially curative gastric cancer: Geographical variations and future prospects. World J Gastroenterol. 2014;20(36):12892–9.CrossRef
3.
Zurück zum Zitat Dai T, Popa E, Shah MA. The role of 18F-FDG PET imaging in upper gastrointestinal malignancies. Curr Treat Options Oncol. 2014;15:351–64.CrossRefPubMed Dai T, Popa E, Shah MA. The role of 18F-FDG PET imaging in upper gastrointestinal malignancies. Curr Treat Options Oncol. 2014;15:351–64.CrossRefPubMed
4.
Zurück zum Zitat Kostakoglu L, Coleman M, Leonard JP, Kuji I, Zoe H, Goldsmith SJ. Positron emission tomography predicts prognosis after one cycle of chemotherapy in aggressive lymphoma and Hodgkin’s disease. J Nucl Med. 2002;43:1018–27.PubMed Kostakoglu L, Coleman M, Leonard JP, Kuji I, Zoe H, Goldsmith SJ. Positron emission tomography predicts prognosis after one cycle of chemotherapy in aggressive lymphoma and Hodgkin’s disease. J Nucl Med. 2002;43:1018–27.PubMed
5.
Zurück zum Zitat Schwarz-Dose J, Untch M, Tiling R, Sassen S, Mahner S, Kahlert S, et al. Monitoring primary systemic therapy of large and locally advanced breast cancer by using sequential positron emission tomography imaging with [18F]fluorodeoxyglucose. J Clin Oncol. 2009;27(4):535–41.CrossRefPubMed Schwarz-Dose J, Untch M, Tiling R, Sassen S, Mahner S, Kahlert S, et al. Monitoring primary systemic therapy of large and locally advanced breast cancer by using sequential positron emission tomography imaging with [18F]fluorodeoxyglucose. J Clin Oncol. 2009;27(4):535–41.CrossRefPubMed
6.
Zurück zum Zitat Avril N, Sassen S, Schmalfeldt B, Naehrig J, Rutke S, Weber WA, et al. Prediction of response to neoadjuvant chemotherapy by sequential F-18-fluorodeoxyglucose positron emission tomography in patients with advanced-stage ovarian cancer. J Clin Oncol. 2005;23(30):7445–53.CrossRefPubMed Avril N, Sassen S, Schmalfeldt B, Naehrig J, Rutke S, Weber WA, et al. Prediction of response to neoadjuvant chemotherapy by sequential F-18-fluorodeoxyglucose positron emission tomography in patients with advanced-stage ovarian cancer. J Clin Oncol. 2005;23(30):7445–53.CrossRefPubMed
7.
Zurück zum Zitat Yoo SW, Kim J, Chong A, Kwon SY, Min JJ, Song HC, et al. Metabolic tumor volume measured by F-18 FDG PET/CT can further stratify the prognosis of patients with Stage IV non-small cell lung cancer. Nucl Med. Mol Imaging. 2012;46:286–96. Yoo SW, Kim J, Chong A, Kwon SY, Min JJ, Song HC, et al. Metabolic tumor volume measured by F-18 FDG PET/CT can further stratify the prognosis of patients with Stage IV non-small cell lung cancer. Nucl Med. Mol Imaging. 2012;46:286–96.
8.
Zurück zum Zitat Becker K, Langer R, Reim D, Novotny A, zum Buschenfelde CM, Engel J, et al. Significance of histopathological tumor regression after neoadjuvant chemotherapy in gastric adenocarcinomas: a summary of 480 cases. Ann Surg. 2011;253(5):934–9.CrossRefPubMed Becker K, Langer R, Reim D, Novotny A, zum Buschenfelde CM, Engel J, et al. Significance of histopathological tumor regression after neoadjuvant chemotherapy in gastric adenocarcinomas: a summary of 480 cases. Ann Surg. 2011;253(5):934–9.CrossRefPubMed
9.
Zurück zum Zitat Langer R, Ott K, Feith M, Lordick F, Siewert JR, Becker K. Prognostic significance of histopathological tumor regression after neoadjuvant chemotherapy in esophageal adenocarcinomas. Mod Pathol. 2009;22(12):1555–63.CrossRefPubMed Langer R, Ott K, Feith M, Lordick F, Siewert JR, Becker K. Prognostic significance of histopathological tumor regression after neoadjuvant chemotherapy in esophageal adenocarcinomas. Mod Pathol. 2009;22(12):1555–63.CrossRefPubMed
10.
Zurück zum Zitat Lordick F, Ott K, Krause BJ, Weber WA, Becker K, Stein HJ, et al. PET to assess early metabolic response and to guide treatment of adenocarcinoma of the oesophagogastric junction: the MUNICON phase II trial. Lancet Oncol. 2007;8(9):797–805.CrossRefPubMed Lordick F, Ott K, Krause BJ, Weber WA, Becker K, Stein HJ, et al. PET to assess early metabolic response and to guide treatment of adenocarcinoma of the oesophagogastric junction: the MUNICON phase II trial. Lancet Oncol. 2007;8(9):797–805.CrossRefPubMed
11.
Zurück zum Zitat Ott K, Fink U, Becker K, Stahl A, Dittler HJ, Busch R, et al. Prediction of response to preoperative chemotherapy in gastric carcinoma by metabolic imaging: results of a prospective trial. J Clin Oncol. 2003;21(24):4604–10.CrossRefPubMed Ott K, Fink U, Becker K, Stahl A, Dittler HJ, Busch R, et al. Prediction of response to preoperative chemotherapy in gastric carcinoma by metabolic imaging: results of a prospective trial. J Clin Oncol. 2003;21(24):4604–10.CrossRefPubMed
12.
Zurück zum Zitat Shah MA, Yeung H, Coit D, Trocola R, Ilson D, Randazzo J, et al. A phase II study of preoperative chemotherapy with irinotecan(CPT) and cisplatin(CIS) for gastric cancer(NCI 5917): FDG-PET/CT predicts patient outcome. J Clin Oncol. 2007;25:4502.CrossRef Shah MA, Yeung H, Coit D, Trocola R, Ilson D, Randazzo J, et al. A phase II study of preoperative chemotherapy with irinotecan(CPT) and cisplatin(CIS) for gastric cancer(NCI 5917): FDG-PET/CT predicts patient outcome. J Clin Oncol. 2007;25:4502.CrossRef
13.
Zurück zum Zitat Roedl JB, Colen RR, Holalkere NS, Fischman AJ, Choi NC, Blake MA. Adenocarcinomas of the esophagus: Response to chemoradiotherapy is associated with decrease of metabolic tumor volume as measured on PET–CT. Comparison to histopathologic and clinical response evaluation. Radiother Oncol. 2008;89:278–86.CrossRefPubMed Roedl JB, Colen RR, Holalkere NS, Fischman AJ, Choi NC, Blake MA. Adenocarcinomas of the esophagus: Response to chemoradiotherapy is associated with decrease of metabolic tumor volume as measured on PET–CT. Comparison to histopathologic and clinical response evaluation. Radiother Oncol. 2008;89:278–86.CrossRefPubMed
14.
Zurück zum Zitat Wieder HA, Brucher BL, Zimmermann F, Becker K, Lordick F, Beer A, et al. Time course of tumor metabolic activity during chemoradiotherapy of esophageal squamous cell carcinoma and response to treatment. J Clin Oncol. 2004;22(5):900–8.CrossRefPubMed Wieder HA, Brucher BL, Zimmermann F, Becker K, Lordick F, Beer A, et al. Time course of tumor metabolic activity during chemoradiotherapy of esophageal squamous cell carcinoma and response to treatment. J Clin Oncol. 2004;22(5):900–8.CrossRefPubMed
15.
Zurück zum Zitat Kwee RM. Prediction of tumor response to neoadjuvant therapy in patients with esophageal cancer with use of 18F FDG PET: a systematic review. Radiology. 2010;254(3):707–17.CrossRefPubMed Kwee RM. Prediction of tumor response to neoadjuvant therapy in patients with esophageal cancer with use of 18F FDG PET: a systematic review. Radiology. 2010;254(3):707–17.CrossRefPubMed
16.
Zurück zum Zitat Konski AA, Cheng JD, Goldberg M, Li T, Maurer A, Yu JQ, et al. Correlation of molecular response as measured by 18-FDG positron emission tomography with outcome after chemoradiotherapy in patients with esophageal carcinoma. Int J Radiat Oncol Biol Phys. 2007;69(2):358–63.CrossRefPubMedPubMedCentral Konski AA, Cheng JD, Goldberg M, Li T, Maurer A, Yu JQ, et al. Correlation of molecular response as measured by 18-FDG positron emission tomography with outcome after chemoradiotherapy in patients with esophageal carcinoma. Int J Radiat Oncol Biol Phys. 2007;69(2):358–63.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Song SY, Kim JH, Ryu JS, Lee GH, Kim SB, Park SI, et al. FDG-PET in the prediction of pathologic response after neoadjuvant chemoradiotherapy in locally advanced, resectable esophageal cancer. Int J Radiat Oncol Biol Phys. 2005;63(4):1053–9.CrossRefPubMed Song SY, Kim JH, Ryu JS, Lee GH, Kim SB, Park SI, et al. FDG-PET in the prediction of pathologic response after neoadjuvant chemoradiotherapy in locally advanced, resectable esophageal cancer. Int J Radiat Oncol Biol Phys. 2005;63(4):1053–9.CrossRefPubMed
18.
Zurück zum Zitat Lee SM, Kim SH, Lee JM, Im SA, Bang YJ, Kim WH, et al. Usefulness of CT volumetry for primary gastric lesions in predicting pathologic response to neoadjuvant chemotherapy in advanced gastric cancer. Abdom Imaging. 2009;34:430–40.CrossRefPubMed Lee SM, Kim SH, Lee JM, Im SA, Bang YJ, Kim WH, et al. Usefulness of CT volumetry for primary gastric lesions in predicting pathologic response to neoadjuvant chemotherapy in advanced gastric cancer. Abdom Imaging. 2009;34:430–40.CrossRefPubMed
19.
Zurück zum Zitat Vallbohmer D, Holscher AH, Dietlein M, Bollschweiler E, Baldus SE, Monig SP, et al. [18F]-Fluorodeoxyglucose-positron emission tomography for the assessment of histopathologic response and prognosis after completion of neoadjuvant chemoradiation in esophageal cancer. Ann Surg. 2009;250(6):888–94.CrossRefPubMed Vallbohmer D, Holscher AH, Dietlein M, Bollschweiler E, Baldus SE, Monig SP, et al. [18F]-Fluorodeoxyglucose-positron emission tomography for the assessment of histopathologic response and prognosis after completion of neoadjuvant chemoradiation in esophageal cancer. Ann Surg. 2009;250(6):888–94.CrossRefPubMed
20.
Zurück zum Zitat Rice TW, Blackstone EH, Rusch VW. 7th edition of the AJCC cancer staging manual: esophagus and esophagogastric junction. Ann Surg Oncol. 2010;17(7):1721–4.CrossRefPubMed Rice TW, Blackstone EH, Rusch VW. 7th edition of the AJCC cancer staging manual: esophagus and esophagogastric junction. Ann Surg Oncol. 2010;17(7):1721–4.CrossRefPubMed
21.
Zurück zum Zitat Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17(12):3077–9.CrossRefPubMed Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17(12):3077–9.CrossRefPubMed
22.
Zurück zum Zitat Chen Y, Awan N, Haveman JW, Apostolou C, Chang DK, Merrett N. Gastric cancer: Australian outcomes of multi-modality treatment with curative intent. ANZ J Surg. 2016;86(5):386–90.CrossRefPubMed Chen Y, Awan N, Haveman JW, Apostolou C, Chang DK, Merrett N. Gastric cancer: Australian outcomes of multi-modality treatment with curative intent. ANZ J Surg. 2016;86(5):386–90.CrossRefPubMed
23.
Zurück zum Zitat Thomson IG, Gotley DC, Barbour AP, Martin I, Jayasuria N, Thomas J, et al. Treatment results of curative gastric resection from a specialist Australian unit: low volume with satisfactory outcomes. Gastric Cancer. 2014;17(1):152–60.CrossRefPubMed Thomson IG, Gotley DC, Barbour AP, Martin I, Jayasuria N, Thomas J, et al. Treatment results of curative gastric resection from a specialist Australian unit: low volume with satisfactory outcomes. Gastric Cancer. 2014;17(1):152–60.CrossRefPubMed
24.
Zurück zum Zitat Kamel E, Hany TF, Burger C, Treyer V, Lonn AHR, von Schulthess GK, et al. CT vs 68Ge attenuation correction in a combined PET/CT system: evaluation of the effect of lowering the CT tube current. Eur J Nucl Med Mol Imaging. 2002;29:346–50.CrossRefPubMed Kamel E, Hany TF, Burger C, Treyer V, Lonn AHR, von Schulthess GK, et al. CT vs 68Ge attenuation correction in a combined PET/CT system: evaluation of the effect of lowering the CT tube current. Eur J Nucl Med Mol Imaging. 2002;29:346–50.CrossRefPubMed
25.
Zurück zum Zitat Nakamoto Y, Osman M, Cohade C, Marshall LT, Links JM, Kohlmyer S, et al. PET/CT: comparison of quantitative tracer uptake between germanium and CT transmission attenuation corrected images. J Nucl Med. 2002;43:1137–43.PubMed Nakamoto Y, Osman M, Cohade C, Marshall LT, Links JM, Kohlmyer S, et al. PET/CT: comparison of quantitative tracer uptake between germanium and CT transmission attenuation corrected images. J Nucl Med. 2002;43:1137–43.PubMed
26.
Zurück zum Zitat Miyata H, Yamasaki M, Takahashi T, Murakami K, Tanaka K, Yukinori K, et al. Determinants of response to neoadjuvant chemotherapy for esophageal cancer using 18F-fluorodeoxiglucose positron emission tomography (18F-FDG-PET). Ann Surg Oncol. 2014;21:575–82.CrossRefPubMed Miyata H, Yamasaki M, Takahashi T, Murakami K, Tanaka K, Yukinori K, et al. Determinants of response to neoadjuvant chemotherapy for esophageal cancer using 18F-fluorodeoxiglucose positron emission tomography (18F-FDG-PET). Ann Surg Oncol. 2014;21:575–82.CrossRefPubMed
27.
Zurück zum Zitat Port JL, Lee PC, Korst RJ, Liss Y, Meherally D, Christos P, et al. Positron emission tomographic scanning predicts survival after induction chemotherapy for esophageal carcinoma. Ann Thorac Surg. 2007;84(2):393–400.CrossRefPubMed Port JL, Lee PC, Korst RJ, Liss Y, Meherally D, Christos P, et al. Positron emission tomographic scanning predicts survival after induction chemotherapy for esophageal carcinoma. Ann Thorac Surg. 2007;84(2):393–400.CrossRefPubMed
28.
Zurück zum Zitat Boellaard R. Need for standardization of 18F-FDG PET/CT for treatment response assessment. Eur J Nucl Med. 2011; 52(12):93 S-100 S. Boellaard R. Need for standardization of 18F-FDG PET/CT for treatment response assessment. Eur J Nucl Med. 2011; 52(12):93 S-100 S.
29.
Zurück zum Zitat Dassen AE, Lips DJ, Hoekstra CJ, Pruijt JFM, Bosscha K. FDG-PET has no definite role in preoperative imaging in gastric cancer. Eur J Surg Oncol. 2009;35(5):449–55.CrossRefPubMed Dassen AE, Lips DJ, Hoekstra CJ, Pruijt JFM, Bosscha K. FDG-PET has no definite role in preoperative imaging in gastric cancer. Eur J Surg Oncol. 2009;35(5):449–55.CrossRefPubMed
30.
Zurück zum Zitat Vallbohmer D, Holscher AH, Schneider PM, Schmidt M, Dietlein M, Bollschweiler E, et al. [18F]-fluorodeoxyglucose-positron emission tomography for the assessment of histopathologic response and prognosis after completion of neoadjuvant chemotherapy in gastric cancer. J Surg Oncol. 2010;102(2):135–40.CrossRefPubMed Vallbohmer D, Holscher AH, Schneider PM, Schmidt M, Dietlein M, Bollschweiler E, et al. [18F]-fluorodeoxyglucose-positron emission tomography for the assessment of histopathologic response and prognosis after completion of neoadjuvant chemotherapy in gastric cancer. J Surg Oncol. 2010;102(2):135–40.CrossRefPubMed
31.
Zurück zum Zitat Chung HW, Lee EJ, Cho YH, Yoon SY, So Y, Kim SY, et al. High FDG uptake in PET/CT predicts worse prognosis in patients with metastatic gastric adenocarcinoma. J Cancer Res Clin Oncol. 2010;136(12):1929–35.CrossRefPubMed Chung HW, Lee EJ, Cho YH, Yoon SY, So Y, Kim SY, et al. High FDG uptake in PET/CT predicts worse prognosis in patients with metastatic gastric adenocarcinoma. J Cancer Res Clin Oncol. 2010;136(12):1929–35.CrossRefPubMed
32.
Zurück zum Zitat Sasaki R, Komaki R, Macapinlac H, Erasmus J, Allen P, Forster K, et al. [18F] Fluorodeoxyglucose uptake by positron emission tomography predicts outcome of non–small-cell lung cancer. J Clin Oncol 2005; 23:1136–1143.CrossRefPubMed Sasaki R, Komaki R, Macapinlac H, Erasmus J, Allen P, Forster K, et al. [18F] Fluorodeoxyglucose uptake by positron emission tomography predicts outcome of non–small-cell lung cancer. J Clin Oncol 2005; 23:1136–1143.CrossRefPubMed
33.
Zurück zum Zitat Tamandl D, Ta J, Schmid R, Preusser M, Paireder M, Schoppmann SF, et al. Prognostic value of volumetric PET parameters in unresectable and metastatic esophageal cancer. Eur J Radiol. 2016;85:540–5.CrossRefPubMed Tamandl D, Ta J, Schmid R, Preusser M, Paireder M, Schoppmann SF, et al. Prognostic value of volumetric PET parameters in unresectable and metastatic esophageal cancer. Eur J Radiol. 2016;85:540–5.CrossRefPubMed
34.
Zurück zum Zitat Grabinska K, Pelak M, Wydmanski J, Tukiendorf A, d’Amico A. Prognostic value and clinical correlations of 18-Fluorodeoxyglucose metabolism quantifiers in gastric cancer. World J Gastroenterol. 2015;21(19):5901–9.PubMedPubMedCentral Grabinska K, Pelak M, Wydmanski J, Tukiendorf A, d’Amico A. Prognostic value and clinical correlations of 18-Fluorodeoxyglucose metabolism quantifiers in gastric cancer. World J Gastroenterol. 2015;21(19):5901–9.PubMedPubMedCentral
35.
Zurück zum Zitat Chen R, Zhou X, Liu J, Huang G. Relationship between 18F-FDG PET/CT findings and HER2 expression in gastric cancer. J Nucl Med. 2016;57:1040–44.CrossRefPubMed Chen R, Zhou X, Liu J, Huang G. Relationship between 18F-FDG PET/CT findings and HER2 expression in gastric cancer. J Nucl Med. 2016;57:1040–44.CrossRefPubMed
36.
Zurück zum Zitat zum Buschenfelde CM, Herrmann K, Schuster T, Geinitz H, Langer R, Becker K, et al. (18)F-FDG PET-guided salvage neoadjuvant radiochemotherapy of adenocarcinoma of the esophagogastric junction: the MUNICON II trial. J Nucl Med. 2011;52(8):1189–96.CrossRefPubMed zum Buschenfelde CM, Herrmann K, Schuster T, Geinitz H, Langer R, Becker K, et al. (18)F-FDG PET-guided salvage neoadjuvant radiochemotherapy of adenocarcinoma of the esophagogastric junction: the MUNICON II trial. J Nucl Med. 2011;52(8):1189–96.CrossRefPubMed
37.
Zurück zum Zitat Won E, Shah MA, Schöder H, Strong VE, Coit DG, Brennan MF, et al. Use of positron emission tomography scan response to guide treatment change for locally advanced gastric cancer: the Memorial Sloan Kettering Cancer Center experience. J Gastrointest Oncol 2016;7(4):506–14. Won E, Shah MA, Schöder H, Strong VE, Coit DG, Brennan MF, et al. Use of positron emission tomography scan response to guide treatment change for locally advanced gastric cancer: the Memorial Sloan Kettering Cancer Center experience. J Gastrointest Oncol 2016;7(4):506–14.
38.
Zurück zum Zitat Hyun SH, Choi JY, Shim YM, Kim K, Lee SJ, Cho YS, et al. Prognostic value of metabolic tumor volume measured by 18F-Fluorodeoxyglucose positron emission tomography in patients with esophageal carcinoma. Ann Surg Oncol. 2010;17(1):115–22.CrossRefPubMed Hyun SH, Choi JY, Shim YM, Kim K, Lee SJ, Cho YS, et al. Prognostic value of metabolic tumor volume measured by 18F-Fluorodeoxyglucose positron emission tomography in patients with esophageal carcinoma. Ann Surg Oncol. 2010;17(1):115–22.CrossRefPubMed
39.
Zurück zum Zitat Coupe NA, Karikios D, Chong S, Yap J, Ng W, Merrett N, et al. Metabolic information on staging FDG-PET–CT as a prognostic tool in the evaluation of 97 patients with gastric cancer. Ann Nucl Med. 2014;28:128–35.CrossRefPubMed Coupe NA, Karikios D, Chong S, Yap J, Ng W, Merrett N, et al. Metabolic information on staging FDG-PET–CT as a prognostic tool in the evaluation of 97 patients with gastric cancer. Ann Nucl Med. 2014;28:128–35.CrossRefPubMed
40.
Zurück zum Zitat Kato H, Takita J, Miyazaki T, Nakajima M, Fukai Y, Masuda N, et al. Correlation of 18-F-fluorodeoxyglucose (FDG) accumulation with glucose transporter (Glut-1) expression in esophageal squamous cell carcinoma. Anticancer Res. 2003;23(4):3263–72.PubMed Kato H, Takita J, Miyazaki T, Nakajima M, Fukai Y, Masuda N, et al. Correlation of 18-F-fluorodeoxyglucose (FDG) accumulation with glucose transporter (Glut-1) expression in esophageal squamous cell carcinoma. Anticancer Res. 2003;23(4):3263–72.PubMed
41.
Zurück zum Zitat Schreurs LMA, Smit JK, Pavlov K, Pultrum BB, Pruim J, Groen H, et al. Prognostic impact of clinicopathological features and expression of biomarkers related to 18F-FDG uptake in esophageal cancer. Ann Surg Oncol. 2014;21(12):3751–7.CrossRefPubMed Schreurs LMA, Smit JK, Pavlov K, Pultrum BB, Pruim J, Groen H, et al. Prognostic impact of clinicopathological features and expression of biomarkers related to 18F-FDG uptake in esophageal cancer. Ann Surg Oncol. 2014;21(12):3751–7.CrossRefPubMed
Metadaten
Titel
Serial imaging using [18F]Fluorodeoxyglucose positron emission tomography and histopathologic assessment in predicting survival in a population of surgically resectable distal oesophageal and gastric adenocarcinoma following neoadjuvant therapy
Publikationsdatum
15.03.2017
Erschienen in
Annals of Nuclear Medicine / Ausgabe 4/2017
Print ISSN: 0914-7187
Elektronische ISSN: 1864-6433
DOI
https://doi.org/10.1007/s12149-017-1159-2

Weitere Artikel der Ausgabe 4/2017

Annals of Nuclear Medicine 4/2017 Zur Ausgabe