Skip to main content
Erschienen in: European Radiology 1/2020

Open Access 07.08.2019 | Gastrointestinal

Gastric cancer and image-derived quantitative parameters: Part 2—a critical review of DCE-MRI and 18F-FDG PET/CT findings

verfasst von: Lei Tang, Xue-Juan Wang, Hideo Baba, Francesco Giganti

Erschienen in: European Radiology | Ausgabe 1/2020

Abstract

There is yet no consensus on the application of functional imaging and qualitative image interpretation in the management of gastric cancer. In this second part, we will discuss the role of image-derived quantitative parameters from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) in gastric cancer, as both techniques have been shown to be promising and useful tools in the clinical decision making of this disease. We will focus on different aspects including aggressiveness assessment, staging and Lauren type discrimination, prognosis prediction and response evaluation. Although both the number of articles and the patients enrolled in the studies were rather small, there is evidence that quantitative parameters from DCE-MRI such as Ktrans, Ve, Kep and AUC could be promising image-derived surrogate parameters for the management of gastric cancer. Data from 18F-FDG PET/CT studies showed that standardised uptake value (SUV) is significantly associated with the aggressiveness, treatment response and prognosis of this disease. Along with the results from diffusion-weighted MRI and contrast-enhanced multidetector computed tomography presented in Part 1 of this critical review, there are additional image-derived quantitative parameters from DCE-MRI and 18F-FDG PET/CT that hold promise as effective tools in the diagnostic pathway of gastric cancer.

Key Points

Quantitative analysis from DCE-MRI and18F-FDG PET/CT allows the extrapolation of multiple image-derived parameters.
Data from DCE-MRI (Ktrans, Ve, Kep and AUC) and 18F-FDG PET/CT (SUV) are non-invasive, quantitative image-derived parameters that hold promise in the evaluation of the aggressiveness, treatment response and prognosis of gastric cancer.
Hinweise
Lei Tang and Xue-Juan Wang contributed equally to this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
18F-FDG PET/CT
18F-Fluorodeoxyglucose positron emission tomography/computed tomography
ADC
Apparent diffusion coefficient
CT
Computed tomography
DCE-MRI
Dynamic contrast-enhanced magnetic resonance imaging
EGFR
Epidermal growth factor receptor
GC
Gastric cancer
SUV
Standardised uptake value
VEGF
Vascular endothelial growth factor
HER
Human epidermal growth factor

Introduction

Gastric cancer (GC) is one of the most common malignancies worldwide [1]. As already discussed in the first part (Part 1) of this critical review [2], this disease is managed through a standardised multidisciplinary approach where radiology plays a crucial role in the detection, staging, treatment planning and follow-up [3, 4].
The most useful techniques are endoscopic ultrasound, computed tomography (CT), magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET)/CT. At this regard, the PLASTIC trial [5] is an ongoing study that will evaluate the impact and cost-effectiveness of PET and staging laparoscopy in addition to initial staging in patients with locally advanced GC.
Different image-derived quantitative parameters from these techniques could be considered promising tools in the management of GC [6, 7], as they reflect a variety of biological processes (normal or pathological) both at baseline and after therapeutic interventions.
Quantitative imaging has the potential to improve the value of diagnostic testing and enhance clinical productivity and is increasingly important in preclinical studies, clinical research, and clinical practice [7]. Oncological imaging represents an ideal setting for the collection of new image-derived quantitative parameters from different techniques that can be potentially included in the clinical scenario [6]. The Radiological Society of North America underlined their importance as non-invasive tools with different applications in oncology and has promoted their use in clinical trials [7].
In the second part, we will provide a critical review on the state of the art of dynamic contrast-enhanced (DCE) MRI and 18F-FDG PET/CT findings.

Evidence acquisition

We searched MEDLINE/PubMed for manuscripts published from inception to 17 August 2018 (Fig. 1).

DCE-MRI and image-derived quantitative parameters

DCE-MRI is a functional imaging technique in which multiphase images are acquired over a few minutes at baseline, during and after rapid intravenous injection of a contrast agent and a saline flush. Changes in signal intensity (reflecting tissue vascularity) can be observed and parametric maps of specific microvascular image-derived quantitative parameters can be derived [8, 9]. Basic recommendations include an adequate spatial/temporal resolution and knowledge of the inherent characteristics of the contrast agent. Semi-quantitative and quantitative analysis can be performed on specific regions of interest (ROIs) or on a pixel-by-pixel basis.
DCE-MRI requires high temporal resolution (usually 4–6 s/phase) and can be degraded by motion artefacts (e.g. respiratory or bowel peristalsis) [10]. Therefore, an injection of intravenous/intramuscular anti-peristaltic agent is advised to reduce the mobility of the gastric walls.
DCE-MRI reflects tumour angiogenesis (i.e. the creation of new blood vessels) and is directly associated with tumour growth and inversely correlated with prognosis [1113].
Different quantitative parameters can be extrapolated from DCE-MRI maps (Tofts model) [14] such as:
  • Ktrans (min−1): volume transfer constant of gadolinium from blood plasma to the extravascular extracellular space (EES)
  • Ve (0 to 100%): volume of the EES per unit volume of tissue (i.e. the amount of “space” available within the interstitium for accumulating gadolinium)
  • Kep (min−1): rate constant gadolinium reflux from the EES back into the vascular system (i.e. it is the ratio: Ktrans/Ve)
  • AUC (mmol/s): area under the gadolinium concentration curve during a certain period of time.
The application of DCE-MRI in GC has been increasingly growing over the last few years thanks to the technical developments (e.g. the shortening of temporal resolution) and the advantage of free-from-radiation damage compared with CT.
Although certainly interesting in a research context, this technique has been mainly applied for neuro-oncological imaging so far. However, DCE-MRI in organ systems outside the central nervous system for oncological applications remains an active area of research, especially for breast, liver and prostate cancer. Other applications of DCE-MRI have been investigated, but as yet are not routinely used in clinical practice for GC. A possible explanation is that tumours are biologically complex structures and, differently from other organs such as the brain, the DCE-MRI protocols for GC are flawed by the presence of several artefacts (especially due to peristalsis) that can easily undermine the quality of the scan and the interpretation of quantitative data from the regions of interest analysed.

DCE-MRI in the detection and diagnosis of gastric cancer

Table 1 summarises the main studies analysing the role of DCE-MRI in GC.
Table 1
Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and gastric cancer
Study (ref.)
Year
Country
Type of study
No. of patients
MRI system
DCE acquisition
ROI placement
Imaging parameter
Key message
Kang et al [15]
2000
South Korea
Prospective
46
1.5 T
Precontrast
30, 60, and 90 s after injection
Delayed scan 4–5 min after injection
Normal and pathologic gastric wall by 2 radiologists in consensus (single slice)
Thickness of the gastric wall
Time to intensity curve (peak enhancement)
Stomach cancer has a thickened wall with rapid enhancement
Pathological mucosa and/or submucosa show early enhancement pattern
Dynamic and delayed MRI can predict preoperative T staging
Joo et al [16]
2014
South Korea
Prospective
27a
3 T
Radial VIBE sequences continuously scanned for 75 s
Repeated
volumetric sets of axial images at 4.1-s intervals for 308 s
Normal and pathologic gastric wall by 1 radiologist (single slice)
Ktrans
Kep
Ve
iAUC (first 60 s)
Ve and iAUC are significantly higher in gastric cancer
Ve is positively correlated with T staging
Ktrans is significantly correlated with EGFR expression
DCE-MRI parameters provide prognostic information for gastric cancer.
Ma et al [17]
2016
China
Prospective
32
3 T
Acquisition time, 15 s
Sequence was repeated 20 times at 10-s intervals
Pathologic gastric wall by 1 radiologist (single slice)
Ktrans
Kep
Ve
iAUC (first 60 s)
Mucinous adenocarcinomas show higher Ve and lower Ktrans.
Diffuse type shows higher Ve than the intestinal type
Mean Ktrans is positively correlated with VEGF
DCE-MRI predicts tumour histological type, Lauren classification and estimation of tumour angiogenesis
Li et al [18]
2017
China
Prospective
43b
3 T
Total acquisition time = 4 min 26 s (FB radial-VIBE) + 20 s for conventional BH VIBE
Normal and pathologic gastric wall by 1 radiologist (single slice)
Ktrans
Kep
Ve
iAUC (first 60 s)
Gastric cancer shows higher Ve and lower Kep
MRI magnetic resonance imaging, DCE dynamic contrast-enhanced, ROI region of interest, s seconds, VIBE volume-interpolated breath-hold examination, Ktrans volume transfer coefficient, Kep reverse reflux rate constant, Ve extracellular extravascular volume fraction, iAUC initial area under the gadolinium concentration curve, EGFR epidermal growth factor receptor, FB free-breathing, BH breath-hold
aBut 22 with DCE-MRI of diagnostic quality
bBut perfusion analysis on 40 patients
The first study by Kang and colleagues dates back to 2000 [15] and reports the usefulness of dynamic and delayed MRI for T staging. The thickness and enhancement pattern of normal and pathological gastric walls were compared in 46 patients through a dynamic protocol including precontrast images and additional acquisitions of 30, 60, 90 and 240–300 s after injection of gadolinium. The pathological outer layers (mucosa and submucosa) showed earlier enhancement (i.e. between 30 and 90 s) than the normal gastric wall in 43/46 patients (93%) and the peak enhancement of the normal gastric wall was > 90 s in 17/46 patients (37%). A reasonable high consistency between MR staging and pathological staging for all T stages was reported (accuracy for T stage, 83%). Such results, although not related to any specific quantitative parameter, show that dynamic MR imaging was already a promising technique for predicting T staging in GC at that time.
Joo and colleagues [16] correlated DCE-MRI parameters with prognostic factors such as pathological T staging and epidermal growth factor receptor (EGFR) expression. Ve and iAUC were significantly higher for GC (0.133 and 5.533 mmol/s, respectively) when compared with normal gastric wall (0.063 and 3.894, respectively) (all p < 0.05). Additionally, Ve was positively correlated with T staging (ρ = 0.483, p = 0.023) and Ktrans was significantly correlated with EGFR expression (ρ = 0.460, p = 0.031). These findings suggest that DCE-MRI reflects tumour biology, providing prognostic information in patients with GC.
Ma and colleagues [17] compared DCE-MRI parameters in different histological subtypes of GC and investigated their correlation with vascular endothelial growth factor (VEGF) expression levels in 32 patients treated with surgical resection. Differently from the other studies, the ROIs were placed only on the lesions and the size was constant for each patient (10 mm). Mucinous adenocarcinomas showed higher Ve (0.491) and lower Ktrans (0.077 min−1) values than non-mucinous tumours (0.288 and 0.274 min−1, respectively) (p < 0.01). Differences were also observed for the Lauren classification, as the diffuse type showed higher Ve and Ktrans (0.466 and 0.249 min−1, respectively) values than the intestinal type (0.253 and 0.183 min−1, respectively) (p < 0.001). Additionally, Ktrans showed a significant correlation with the level of VEGF expression (ρ = 0.762, p < 0.001). Ktrans and VEGF are both related to the endothelial and microvascular permeability, which are in turn related to the neo-angiogenesis that is seen in tumours: in other words, a higher Ktrans is related to a higher level of VEGF, which is strictly related to a greater degree of angiogenesis. Together with the previous study [16], these findings suggest that angiogenesis increases the extravasation of gadolinium from the intravascular to the interstitial space, supporting the role of DCE-MRI as a potential tool to differentiate GC according to different histopathological features.
Li and colleagues [18] compared the performance of conventional breath-hold to free-breathing DCE-MRI using volume-interpolated breath-hold examination sequences. DCE-MRI parameters of normal gastric wall and GC were collected and perfusion parameters for both normal and pathological gastric walls were obtained. Kep was lower (0.750 vs 1.081 min−1; p < 0.05) while Ve was higher in GC (0.228 vs 0.162; p < 0.05). No significant differences for Ktrans and iAUC values between normal and pathological gastric walls were observed (p > 0.05).
Some examples of DCE-MRI in GC are shown in Figs. 2, 3 and 4.

18F-FDG PET/CT and image-derived quantitative parameters

18F-FDG PET/CT is recommended for patients with newly diagnosed GC if clinically indicated and if metastatic cancer is not evident, as well as in the posttreatment assessment and restaging.
The standardised uptake value (SUV) from 18F-FDG PET/CT is a dimensionless ratio used to distinguish between normal and abnormal levels of glucose uptake and can be considered an image-derived semi-quantitative parameter, defined as the ratio activity per unit volume of a ROI to the activity per unit whole-body volume (Figs. 5 and 6) [19].

18F-FDG PET/CT to assess the primary lesion in gastric cancer

Table 2 summarises the studies on the role of 18F-FDG PET/CT to assess the primary lesion in GC.
Table 2
18F-Fluorodeoxyglucose positron emission tomography (18F-FDG PET) and aggressiveness in gastric cancer
Study (ref.)
Year
Country
Type of study
No. of patients
ROI placement
SUV cut-off
Reference standard
Key messages
Stahl et al [20]
2002
Germany
Prospective
40 (+ 10 controls)
Tumour and normal gastric wall
4.6
Biopsy
18F-FDG PET detected 24/40 (60%) of locally advanced gastric cancers
The mean SUV was higher in the intestinal type than in the non-intestinal type (6.7 vs 4.8; p = 0.03)
The survival rate of patients (n = 36) with 18F-FDG accumulation did not differ from those with low 18F-FDG accumulation (p = 0.75)
Mochiki et al [21]
2004
Japan
Prospective
156
Tumour, lymph nodes and normal gastric wall
4
Radical surgery
Significant association between SUV and the tumour invasion, size and nodal metastasis
18F-FDG PET is less accurate than CT in nodal staging (sensitivity, 23% vs. 65%, respectively)
Survival rate for SUV > 4 was lower than for SUV < 4 (p < 0.05)
18F-FDG PET is not feasible for detecting early-stage gastric cancers
Chen et al [22]
2005
South Korea
Prospective
68
Tumour
Three-point scale: 1 (normal), 2 (equivocal) and 3 (abnormal)a
Radical surgery
18F-FDG PET sensitivity was 94% in patients with gastric cancer
Significant association between 18F-FDG uptake and tumour size, nodal involvement and other histological features
18F-FDG PET + CT is more accurate for preoperative staging than either modality alone (66% vs. 51% and 66% vs. 47%; p = 0.002)
Oh et al [23]
2011
South Korea
Retrospective
136
Tumour
3.2
Radical surgery
SUV was significantly associated with tumour size, depth of invasion and nodal metastasis (p < 0.001) but not with tumour histology (p = 0.099)
Oh et al [24]
2012
South Korea
Retrospective
38
Tumour
Measurable disease was defined as 1.35*SUVmax of liver+2*standard deviation of liver SUV
Radical surgery
31/38 (82%) of tumours were visible on 18F-FDG PET
Measurable tumours on 18F-FDG PET were more frequent in well- or moderately differentiated gastric cancer (p < 0.05), antrum or angle and intestinal type (p > =0.05)
Namikawa et al [25]
2013
Japan
Retrospective
90
NR
NR
Radical surgery
18F-FDG PET CT sensitivity for gastric cancer was 79%
Median SUVmax was significantly different in patients with T3/T4 disease, distant metastasis and stage III/IV tumours
The SUVmax was correlated with tumour size (r = 0.461; p < 0.001)
ROI region of interest, SUV standardised uptake value, PET positron emission tomography, FDG fluorodeoxyglucose, CT computed tomography
a2 and 3 were considered positive
Stahl and colleagues [20] analysed the relationship between SUVmean and different tumour features from biopsy (including intestinal vs non-intestinal) in 40 patients. PET had a sensitivity of 60% in identifying locally advanced GC and the SUVmean was higher in the intestinal than in the non-intestinal type (6.7 vs 4.8; p = 0.03). No significant differences in the survival rate of patients with or without FDG accumulation (SUVmean cut-off, 4.6; p = 0.75) were observed. A clear limitation of this study is that the reference standard was biopsy and not radical surgery.
Mochiki and colleagues [21] reported a significant association between SUVmean and the depth of invasion, tumour size and nodal metastasis. They compared 18F-FDG PET findings with CT and found that 18F-FDG PET was less accurate for nodal staging (23% vs 65%). The SUVmean was higher for T2–T4 than T1 tumours (p < 0.05). Differently from the previous study [20], they observed a significant difference in the survival rate (p < 0.05).
Chen and colleagues [22] reported a sensitivity of 94% for 18F-FDG PET/CT (SUVmean = 7) and a significant association between FDG uptake and tumour size, nodal involvement and other histological features. They were among the first showing that the combination of 18F-FDG PET and CT was more accurate for preoperative staging than either modality alone (66% vs 51%, 66% vs. 47%; p = 0.002).
Oh and colleagues [23] performed a retrospective 18F-FDG PET/CT analysis of 136 patients treated with radical surgery. They set a threshold for SUVpeak from primary tumour of 3.2 to define hypermetabolic lesions and found that this was associated with tumour depth and nodal involvement (p < 0.001). The sensitivity and specificity for nodal involvement using the aforementioned threshold were 75% and 74% respectively.
Another group [24] reported the relationship between measurable and non-measurable GC on 18F-FDG PET/CT (defined as 1.35*SUVmax of liver+2*standard deviation of liver SUV). Among different parameters, a higher proportion of measurable tumours was found in well- or moderately differentiated GC than poorly differentiated tumours (71% vs 33% p < 0.05). Differently from the previous study [24], there was no difference for primary tumour stage and nodal metastasis.
Namikawa and colleagues [25] reported a sensitivity of 79% for the detection of GC for 18F-FDG PET/CT and a significant difference for SUVmax for patients with T3/T4 vs T1/T2 (9.0 vs. 3.8; p < 0.001), with and without distant metastasis (9.5 vs. 7.7; p = 0.018), and between stage III/IV and stage I/II (9.0 vs. 4.7; p = 0.017) after radical surgery. The SUVmax of the primary tumour was correlated with tumour size (r = 0.461; p < 0.001). The sensitivity, specificity and accuracy of 18F-FDG PET/CT for nodal involvement were 64%, 86% and 71% respectively.

18F-FDG PET/CT in treatment response of gastric cancer

We found six studies reporting on 18F-FDG PET/CT and treatment response in GC (Table 3).
Table 3
Fluorodeoxyglucose positron emission tomography (18F-FDG PET) and treatment response in gastric cancer
Study (ref.)
Year
Country
Type of study
No. of patients
ROI placement
SUV reduction to distinguish between responders and non responders
Number of 18F-FDG PET scans
Histological definition of treatment response
Reference standard
Key messages
Stahl et al [26]
2004
Germany
Retrospective
43
Tumour
40%
Baseline and during the first cycle of chemotherapy
< 10% viable tumour cells in the specimen
Surgery
Pretreatment SUV was higher for responders than non-responders (p = 0.09)
SUV after the first cycle of chemotherapy was lower for responders than non-responders (p = 0.36)
SUV changes were significantly higher in responders than non-responders (p < 0.01)
Importance of protocol standardisation
Vallböhmer et al [27]
2013
Germany
Prospective
40
Tumour
NR
Baseline and 2 weeks after completion of chemotherapy
< 10% viable tumour cells in the specimen
Surgery
Overall, posttreatment SUV was significantly lower than pretreatment SUV (p = 0.0006)
No significant correlations between pre- and posttreatment
SUV (and relative changes) and histological treatment response
Higher pretreatment SUV for intestinal (7.8) than diffuse (5.1) types (p = 0.023)
SUV change was significantly different according to tumour location (p = 0.041).
Giganti et al [28]
2014
Italy
Prospective
17
Tumour
NR
Baseline and 2 weeks after completion of chemotherapy
TRG 1–3 were considered responders and TRG 4–5 non-responders
Surgery
No correlations between pre- or posttreatment SUV (and % change) and treatment response
Wang et al [29]
2015
China
Prospective
64
Tumour + metastatic sites (liver, nodes and ovary)
40% (primary tumour)
Baseline and 14 days after start of chemotherapy
NRa
Imaging (unresectable gastric cancer)
A 40% uptake reduction is the cut-off to predict clinical response (sensitivity of 70% and specificity of 83%) to predict
Early metabolic change might be a predictive marker for response and disease control in advanced gastric cancer
Park et al [30]
2016
South Korea
Prospective
74
Tumour
50%
Baseline and 6 weeks after start of chemotherapy
NR
Imaging (unresectable gastric cancer)
A 50% SUVmax reduction was associated with a 30% tumour size reduction (p < 0.001)
Poorly cohesive carcinomas demonstrate lower
SUVmax irrespective of tumour size (p < 0.001)
HER2–positive tumours showed increased SUVmax than HER2–negative tumours (p = 0.002)
Schneider et al [31]
2018
Switzerland
Retrospective
30
Tumour
35%
Baseline and 2 weeks after the completion of chemotherapy
< 10% viable tumour cells in the specimen
Surgery
Metabolic response was observed in 67% and no response in 33%
Prediction of pathological response by SUV had a sensitivity of 91% and a specificity of 47%, with an overall accuracy of 63%
ROI region of interest, SUV standardised uptake value, PET positron emission tomography, NR not reported, TRG tumour regression grade, HER human epidermal growth factor receptor
aRECIST criteria were used
Stahl and colleagues [26] compared different 18F-FDG PET/CT protocols and calculations of the SUVmean (time delay after 18F-FDG administration, acquisition protocol, reconstruction algorithm, SUV normalisation) for the early prediction of treatment response at baseline and after the first cycle of chemotherapy. They did not find any significant difference in the baseline and follow-up SUVmean calculation between protocols (p > 0.05), but higher SUV changes for responders than non-responders were observed (p < 0.01). They were among the first to demonstrate the robustness of 18F-FDG PET/CT for therapeutic monitoring, supporting the comparability of studies obtained with different protocols.
Vallböhmer and colleagues [27] analysed the differences in pre- and posttreatment SUVmax between responders and non-responders using the same histological definition as Stahl [26] (i.e. < 10% viable tumour cells in the specimen) but no correlation with treatment response was observed (p = 0.733). Significant differences in SUVmax were observed for the Lauren classification (p = 0.023) and tumour location (p = 0.041).
In another study on 17 patients [28] undergoing diffusion-weighted MRI and 18F-FDG PET/CT before and after treatment, no differences in treatment response were observed for pre- or posttreatment SUVmean (and their percentage change) (p = 0.605, p = 0.524 and p = 0.480). Treatment response was based on tumour regression grade (TRG) [32] and responders were considered TRG 1, 2 and 3 (i.e. including patients with more than 10% of viable cells).
Two studies [29, 30] evaluated the relationship between SUVmax and treatment response in advanced GC (i.e. no surgical specimens were used as the reference standard). Although follow-up imaging was performed at different time points (14 days vs 6 weeks after the start of chemotherapy) and different SUV thresholds for response were applied (40% vs 50%), both studies showed that metabolic changes in 18F-FDG PET/CT are predictive markers for response disease also for advanced GC. One study [30] showed a correlation between human epidermal growth factor HER2 status positivity (i.e. more aggressive cancer) and higher SUV uptake (p = 0.002).
Schneider and colleagues [31] reported that 18F-FDG PET/CT is able to detect non-responders (sensitivity, 91%; specificity, 47%; positive predictive value, 50%; negative predictive value, 90%; accuracy, 63%) but they could not prove that 18F-FDG PET/CT after the first cycle of chemotherapy can predict overall pathological response.
Similarly to the PRIDE study in oesophageal cancer [33], there is growing interest to develop models that predict the probability of response to neoadjuvant therapy in GC based on quantitative parameters derived from MRI and 18F-FDG PET/CT. However, given the controversial results at this regard [34], further studies are needed.

18F-FDG PET/CT in the prognosis of gastric cancer

We found eight studies on 18F-FDG PET/CT and prognosis in GC (Table 4). Significant results on the relationship between SUVmax and SUVmean and overall survival were reported by seven of them [3538, 4042], even though each study used different SUVmax and SUV mean cut-offs (Table 4). The study that did not show any significant difference in SUVmax and SUVmean with regard to prognosis was performed by Grabinska and colleagues [39]. A possible explanation is that a long range of follow-up was introduced in this study (range, 6 days to 5.2 years; median, 9.5 months), as also reported by the same authors. Therefore, the survival analysis from their study should be interpreted with caution. However, there is evidence of the relationship between SUVmax and SUVmean and prognosis in GC (Table 4).
Table 4
18F-Fluorodeoxyglucose positron emission tomography (18F-FDG PET) and prognosis in gastric cancer
Study (ref.)
Year
Country
Type of study
No. of patients
Follow-up (months)
ROI placement
SUV cut-off for stomach
Reference standard
Key message
Pak et al [35]
2011
South Korea
NR
41
31
Tumour
3.80
Surgery
The high-SUV group showed more aggressive tumour behaviour in relation to TNM stages (p = 0.018) and more postoperative recurrence (p = 0.028), shorter relapse-free survival (p = 0.004), and lower 30-month cancer-specific survival rates (40% vs. 69.3%; p = 0.008)
SUV is not an independent predictor of overall survival at multivariate analysis
Park et al [36]
2012
South Korea
NR
82
NR
Tumour, lymph nodes and other metastatic sites
6
Biopsy
Longer median progression-free survival (8.7 vs. 4.8 months; p = 0.001) and overall survival (15.4 vs. 11.2 months; p = 0.006) were observed for patients with SUV < 6
Among patients with histologically undifferentiated carcinomas, those with SUV < 6 showed longer median progression-survival (p = 0.005) and overall survival (p < 0.001)
SUV was as an independent predictor of progression-free survival (p = 0.002) and overall survival (p = 0.038)
Lee et al [37]
2012
South Korea
Retrospective
271
24
Tumour
8.2
Surgery
Tumour size, depth of invasion, nodal involvement, positive 18F-FDG uptake and SUV were significantly associated with tumour recurrence at univariate analysis (p ≤ 0.001)
Depth of invasion, positive 18F-FDG uptake and SUV were significantly different at multivariate analysis (p < 0.005)
The 24-month recurrence-free survival rate was significantly
higher in patients with a negative than in those with a positive 18F-FDG uptake (95% vs 74%; p < 0.0001)
Kim et al [38]
2014
South Korea
Retrospective
97
30
Tumour
5.74
Surgery
Progression-free survival of the group with SUV ≤ 5.74 was significantly longer (30.9 months) than that with SUV > 5.74 (24.3 months) (p = 0.008)
In multivariate analysis, high SUV (> 5.74) is the only poor prognostic factor for progression-free survival (p = 0.002; HR = 11.03)
Grabinska et al [39]
2015
Poland
Retrospective
40
9.5
Tumour
NR for prognosis
Biopsy
Despite a difference in median SUV between confined and disseminated gastric cancer (10.36 vs 12.78), no significant difference in SUV was observed with regard to prognosis
Na et al [40]
2016
South Korea
Retrospective
133
43
Tumour
4.3
Surgery
Patients with higher SUV had shorter overall survival (p = 0.008) at univariate analysis but not after adjusting for other clinical parameters (p = 0.28)
SUV was significantly associated with shorter recurrence-free survival (p = 0.003), but not after adjusting for other clinical factors (p = 0.06)
Lee et al [41]
2017
South Korea
Retrospective
44
44
Tumour
1.45a
Biopsy/surgery
The overall survival for patients with SUV > 1.45 was not significantly different (p = 0.068) at univariate analysis but it was at multivariate analysis (HR, 2.026; p = 0.054)
The progression-free survival for patients with SUV > 1.45 was significantly different both at univariate (p = 0.046) and multivariate analyses (HR, 2.105; p = 0.036)
Chon et al [42]
2018
South Korea
Retrospective
727
32.5
Tumour
7.6b
4.6c
5.6d
Surgery
In multivariate analysis, high SUV was negatively correlated with disease-free survival (HR, 2.17) and overall survival (HR, 2.47) (both p < 0.001) in patients with diffuse type
In multivariate analysis, high SUV was negatively correlated with disease-free survival (HR, 2.26; p = 0.005) and overall survival (HR, 2.61; p = 0.003) in patients with signet ring cell carcinoma
This negative prognostic impact was not observed in patients with intestinal type or well- or moderately differentiated histology
ROI region of interest, NR not reported, SUV standardised uptake value, TNM tumour node metastasis, 18F-FDG 18-fluorodeoxyglucose, HR hazard ratio
aAfter chemotherapy
bIntestinal type
cDiffuse type
dMixed type

18F-FDG PET/CT and radiomics in gastric cancer

There is growing evidence of the importance of radiomics in medical imaging [43] and this applies also to 18F-FDG PET/CT findings [44, 45].
A recent review has shown the promising role of radiomics obtained from different techniques—including 18F-FDG PET/CT—in gastro-oesophageal tumours [46].
Jiang and colleagues [47] have also developed a dedicated radiomic score using the features from 18F-FDG PET/CT in GC. In their study, they concluded that the radiomic signature was a powerful predictor of overall and disease-free survival and could add prognostic value to the traditional staging system.
However, as the current literature on this specific topic is still preliminary, there is a need of standardisation and different multicentre studies before including radiomics from 18F-FDG PET/CT in the clinical routine for GC.

Limitations

Quantitative imaging is becoming an increasingly common tool in modern radiology and its potential impact on patient care and on clinical outcomes is huge. However, it is broadly accepted that surrogate quantitative parameters of tumour biology assessed by imaging still require extensive standardisation and validation to proof that the surrogate represents the pathophysiological process under investigation. As reported by Rosenkrantz and colleagues [48], there are some practical aspects that should be considered when discussing the role of image-derived quantitative parameters. These are (i) accuracy (of a measurement, for example); (ii) repeatability and (iii) reproducibility (especially when quantitative imaging is performed in serial scans over time, as this allows to discriminate measurement error from biologic change) and (iv) clinical validity (i.e. impacting and improving patient’s life).
Therefore, some limitations from the papers discussed in this study should be reported. Firstly, for DCE-MRI, our review shows that the ROIs in all studies have been drawn on one selected axial section. This represents an important limitation, as these findings may be less representative of the whole tumour. Future studies should perform quantitative analysis on the whole volume obtained by contouring the tumour borders on each slice by planimetry. There is also a lack of optimised perfusion MRI protocols, dedicated postprocessing software programmes and high variability between MR scanners.
As far as 18F-FDG PET/CT imaging is concerned, a clear limitation is that the SUV is dependent on many factors including the ROI delineation, the activity injected, plasma glucose levels, and body size. There is variability between 18F-FDG PET/CT scanners, as well as in the accuracy of the image reconstruction and correction algorithms. The increased 18F-FDG uptake can be also seen in inflammatory or granulomatous processes and in sites of physiological tracer biodistribution.
Gastric distention, achieved by the consumption of water, milk or foaming agents before scanning, and a late-time-point 18F-FDG PET/CT scanning can relatively differentiate the physiological uptake from the malignant lesion.
Finally, standardised guidelines on how to interpret the quantitative results from DCE-MRI and 18F-FDG PET/CT have yet to be reported.

Conclusions

Similarly to the ADC from diffusion-weighted MRI and texture analysis from CT [2], different image-derived quantitative parameters from DCE-MRI and 18F-FDG PET/CT are promising tools in the management of GC. However, extensive standardisation and validation are still required before they can become an essential cornerstone for GC.

Compliance with ethical standards

Guarantor

The scientific guarantor of this publication is Dr. Francesco Giganti.

Conflict of interest

The authors of this manuscript declare no relationships with any companies, whose products or services may be related to the subject matter of the article.

Statistics and biometry

No complex statistical methods were necessary for this paper.
Written informed consent was not required for this study.

Ethical approval

Institutional Review Board approval was not required.

Methodology

• Review
• Multicentre study
Open Access This 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.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

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.

Jetzt bestellen und 100 € sparen!

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 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D (2011) Global cancer statistics. CA Cancer J Clin 61(2):69–90CrossRef Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D (2011) Global cancer statistics. CA Cancer J Clin 61(2):69–90CrossRef
3.
Zurück zum Zitat Giganti F, Orsenigo E, Arcidiacono PG et al (2016) Preoperative locoregional staging of gastric cancer: is there a place for magnetic resonance imaging? Prospective comparison with EUS and multidetector computed tomography. Gastric Cancer 19(1):216–225CrossRef Giganti F, Orsenigo E, Arcidiacono PG et al (2016) Preoperative locoregional staging of gastric cancer: is there a place for magnetic resonance imaging? Prospective comparison with EUS and multidetector computed tomography. Gastric Cancer 19(1):216–225CrossRef
4.
Zurück zum Zitat Richman DM, Tirumani SH, Hornick JL et al (2017) Beyond gastric adenocarcinoma: multimodality assessment of common and uncommon gastric neoplasms. Abdom Radiol (NY) 42(1):124–140 Richman DM, Tirumani SH, Hornick JL et al (2017) Beyond gastric adenocarcinoma: multimodality assessment of common and uncommon gastric neoplasms. Abdom Radiol (NY) 42(1):124–140
5.
Zurück zum Zitat Brenkman HJF, Gertsen EC, Vegt E et al (2018) Evaluation of PET and laparoscopy in STagIng advanced gastric cancer: a multicenter prospective study (PLASTIC-study). BMC Cancer 18(1):450CrossRef Brenkman HJF, Gertsen EC, Vegt E et al (2018) Evaluation of PET and laparoscopy in STagIng advanced gastric cancer: a multicenter prospective study (PLASTIC-study). BMC Cancer 18(1):450CrossRef
6.
Zurück zum Zitat European Society of Radiology (ESR) (2010) White paper on imaging biomarkers. Insights Imaging 1(2):42–45CrossRef European Society of Radiology (ESR) (2010) White paper on imaging biomarkers. Insights Imaging 1(2):42–45CrossRef
7.
Zurück zum Zitat Buckler AJ, Bresolin L, Dunnick NR, Sullivan DC (2011) A collaborative enterprise for multi-stakeholder participation in the advancement of quantitative imaging. Radiology 258(3):906–914CrossRef Buckler AJ, Bresolin L, Dunnick NR, Sullivan DC (2011) A collaborative enterprise for multi-stakeholder participation in the advancement of quantitative imaging. Radiology 258(3):906–914CrossRef
8.
Zurück zum Zitat Tofts PS (1997) Modeling tracer kinetics in dynamic Gd-DTPA MR imaging. J Magn Reson Imaging 7(1):91–101CrossRef Tofts PS (1997) Modeling tracer kinetics in dynamic Gd-DTPA MR imaging. J Magn Reson Imaging 7(1):91–101CrossRef
9.
Zurück zum Zitat O’Connor JP, Tofts PS, Miles KA, Parkes LM, Thompson G, Jackson A (2011) Dynamic contrast-enhanced imaging techniques: CT and MRI. Br J Radiol 84(special_issue_2):S112–S120 O’Connor JP, Tofts PS, Miles KA, Parkes LM, Thompson G, Jackson A (2011) Dynamic contrast-enhanced imaging techniques: CT and MRI. Br J Radiol 84(special_issue_2):S112–S120
10.
Zurück zum Zitat Kershaw LE, Cheng HLM (2010) Temporal resolution and SNR requirements for accurate DCE-MRI data analysis using the AATH model. Magn Reson Med 64(6):1772–1780CrossRef Kershaw LE, Cheng HLM (2010) Temporal resolution and SNR requirements for accurate DCE-MRI data analysis using the AATH model. Magn Reson Med 64(6):1772–1780CrossRef
11.
Zurück zum Zitat Nishida N, Yano H, Nishida T, Kamura T, Kojiro M (2006) Angiogenesis in cancer. Vasc Health Risk Manag 2(3):213–219CrossRef Nishida N, Yano H, Nishida T, Kamura T, Kojiro M (2006) Angiogenesis in cancer. Vasc Health Risk Manag 2(3):213–219CrossRef
12.
Zurück zum Zitat Tonini T, Rossi F, Claudio PP (2003) Molecular basis of angiogenesis and cancer. Oncogene 22(42):6549–6556CrossRef Tonini T, Rossi F, Claudio PP (2003) Molecular basis of angiogenesis and cancer. Oncogene 22(42):6549–6556CrossRef
13.
Zurück zum Zitat Cuenod CA, Balvay D (2013) Perfusion and vascular permeability: basic concepts and measurement in DCE-CT and DCE-MRI. Diagn Interv Imaging 94(12):1187–1204CrossRef Cuenod CA, Balvay D (2013) Perfusion and vascular permeability: basic concepts and measurement in DCE-CT and DCE-MRI. Diagn Interv Imaging 94(12):1187–1204CrossRef
14.
Zurück zum Zitat Tofts PS, Brix G, Buckley DL et al (1999) Estimating kinetic parameters from dynamic contrast-enhanced T1-weighted MRI of a diffusable tracer: standardized quantities and symbols. J Magn Reson Imaging 10:223–232CrossRef Tofts PS, Brix G, Buckley DL et al (1999) Estimating kinetic parameters from dynamic contrast-enhanced T1-weighted MRI of a diffusable tracer: standardized quantities and symbols. J Magn Reson Imaging 10:223–232CrossRef
15.
Zurück zum Zitat Kang BC, Kim JH, Kim KW et al (2000) Abdominal imaging value of the dynamic and delayed MR sequence with Gd-DTPA in the T-staging of stomach cancer: correlation with the histopathology. Abdom Imaging 25:14–24CrossRef Kang BC, Kim JH, Kim KW et al (2000) Abdominal imaging value of the dynamic and delayed MR sequence with Gd-DTPA in the T-staging of stomach cancer: correlation with the histopathology. Abdom Imaging 25:14–24CrossRef
16.
Zurück zum Zitat Joo I, Lee JM, Han JK, Yang HK, Lee HJ, Choi BI (2015) Dynamic contrast-enhanced MRI of gastric cancer: correlation of the perfusion parameters with pathological prognostic factors. J Magn Reson Imaging 41(6):1608–1614CrossRef Joo I, Lee JM, Han JK, Yang HK, Lee HJ, Choi BI (2015) Dynamic contrast-enhanced MRI of gastric cancer: correlation of the perfusion parameters with pathological prognostic factors. J Magn Reson Imaging 41(6):1608–1614CrossRef
17.
Zurück zum Zitat Ma L, Xu X, Zhang M et al (2017) Dynamic contrast-enhanced MRI of gastric cancer: correlations of the pharmacokinetic parameters with histological type, Lauren classification, and angiogenesis. Magn Reson Imaging 37:27–32CrossRef Ma L, Xu X, Zhang M et al (2017) Dynamic contrast-enhanced MRI of gastric cancer: correlations of the pharmacokinetic parameters with histological type, Lauren classification, and angiogenesis. Magn Reson Imaging 37:27–32CrossRef
18.
Zurück zum Zitat Li HH, Zhu H, Yue L et al (2018) Feasibility of free-breathing dynamic contrast-enhanced MRI of gastric cancer using a golden-angle radial stack-of-stars VIBE sequence: comparison with the conventional contrast-enhanced breath-hold 3D VIBE sequence. Eur Radiol 28(5):1891–1899CrossRef Li HH, Zhu H, Yue L et al (2018) Feasibility of free-breathing dynamic contrast-enhanced MRI of gastric cancer using a golden-angle radial stack-of-stars VIBE sequence: comparison with the conventional contrast-enhanced breath-hold 3D VIBE sequence. Eur Radiol 28(5):1891–1899CrossRef
19.
Zurück zum Zitat Thie JA (2004) Understanding the standardized uptake value, its methods, and implications for usage. J Nucl Med 45(9):1431–1434PubMed Thie JA (2004) Understanding the standardized uptake value, its methods, and implications for usage. J Nucl Med 45(9):1431–1434PubMed
20.
Zurück zum Zitat Stahl A, Ott K, Weber WA et al (2003) FDG PET imaging of locally advanced gastric carcinomas: correlation with endoscopic and histopathological findings. Eur J Nucl Med Mol Imaging 30(2):288–295CrossRef Stahl A, Ott K, Weber WA et al (2003) FDG PET imaging of locally advanced gastric carcinomas: correlation with endoscopic and histopathological findings. Eur J Nucl Med Mol Imaging 30(2):288–295CrossRef
21.
Zurück zum Zitat Mochiki E, Kuwano H, Katoh H, Asao T, Oriuchi N, Endo K (2004) Evaluation of 18F-2-deoxy-2-fluoro-D-glucose positron emission tomography for gastric cancer. World J Surg 28(3):247–253CrossRef Mochiki E, Kuwano H, Katoh H, Asao T, Oriuchi N, Endo K (2004) Evaluation of 18F-2-deoxy-2-fluoro-D-glucose positron emission tomography for gastric cancer. World J Surg 28(3):247–253CrossRef
22.
Zurück zum Zitat Chen J, Cheong JH, Yun MJ et al (2005) Improvement in preoperative staging of gastric adenocarcinoma with positron emission tomography. Cancer 103(11):2383–2390 Chen J, Cheong JH, Yun MJ et al (2005) Improvement in preoperative staging of gastric adenocarcinoma with positron emission tomography. Cancer 103(11):2383–2390
23.
Zurück zum Zitat Oh HH, Lee SE, Choi IS et al (2011) The peak-standardized uptake value (P-SUV) by preoperative positron emission tomography-computed tomography (PET-CT) is a useful indicator of lymph node metastasis in gastric cancer. J Surg Oncol 104(5):530–533CrossRef Oh HH, Lee SE, Choi IS et al (2011) The peak-standardized uptake value (P-SUV) by preoperative positron emission tomography-computed tomography (PET-CT) is a useful indicator of lymph node metastasis in gastric cancer. J Surg Oncol 104(5):530–533CrossRef
24.
Zurück zum Zitat Oh SY, Cheon GJ, Kim YC, Jeong E, Kim S, Choe JG (2012) Detectability of T-measurable diseases in advanced gastric cancer on FDG PET-CT. Nucl Med Mol Imaging 46(4):261–268CrossRef Oh SY, Cheon GJ, Kim YC, Jeong E, Kim S, Choe JG (2012) Detectability of T-measurable diseases in advanced gastric cancer on FDG PET-CT. Nucl Med Mol Imaging 46(4):261–268CrossRef
25.
Zurück zum Zitat Namikawa T, Okabayshi T, Nogami M, Ogawa Y, Kobayashi M, Hanazaki K (2014) Assessment of 18F-fluorodeoxyglucose positron emission tomography combined with computed tomography in the preoperative management of patients with gastric cancer. Int J Clin Oncol 19(4):649–655CrossRef Namikawa T, Okabayshi T, Nogami M, Ogawa Y, Kobayashi M, Hanazaki K (2014) Assessment of 18F-fluorodeoxyglucose positron emission tomography combined with computed tomography in the preoperative management of patients with gastric cancer. Int J Clin Oncol 19(4):649–655CrossRef
26.
Zurück zum Zitat Stahl A, Ott K, Schwaiger M, Weber WA (2004) Comparison of different SUV-based methods for monitoring cytotoxic therapy with FDG PET. Eur J Nucl Med Mol Imaging 31(11):1471–1479CrossRef Stahl A, Ott K, Schwaiger M, Weber WA (2004) Comparison of different SUV-based methods for monitoring cytotoxic therapy with FDG PET. Eur J Nucl Med Mol Imaging 31(11):1471–1479CrossRef
27.
Zurück zum Zitat Vallböhmer D, Hölscher AH, Schneider PM et al (2010) [18F]-Fluorodeoxyglucose-positron emission tomography for the assessment of histopathologic response and prognosis after completion of neoadjuvant chemotherapy in gastric cancer. J Surg Oncol 102(2):135–140 Vallböhmer D, Hölscher AH, Schneider PM et al (2010) [18F]-Fluorodeoxyglucose-positron emission tomography for the assessment of histopathologic response and prognosis after completion of neoadjuvant chemotherapy in gastric cancer. J Surg Oncol 102(2):135–140
28.
Zurück zum Zitat Giganti F, De Cobelli F, Canevari C et al (2014) Response to chemotherapy in gastric adenocarcinoma with diffusion-weighted MRI and 18 F-FDG-PET/CT: correlation of apparent diffusion coefficient and partial volume corrected standardized uptake value with histological tumor regression grade. J Magn Reson Imaging 40(5):1147–1157CrossRef Giganti F, De Cobelli F, Canevari C et al (2014) Response to chemotherapy in gastric adenocarcinoma with diffusion-weighted MRI and 18 F-FDG-PET/CT: correlation of apparent diffusion coefficient and partial volume corrected standardized uptake value with histological tumor regression grade. J Magn Reson Imaging 40(5):1147–1157CrossRef
29.
Zurück zum Zitat Wang C, Guo W, Zhou M et al (2016) The predictive and prognostic value of early metabolic response assessed by positron emission tomography in advanced gastric cancer treated with chemotherapy. Clin Cancer Res 22(7):1603–1610CrossRef Wang C, Guo W, Zhou M et al (2016) The predictive and prognostic value of early metabolic response assessed by positron emission tomography in advanced gastric cancer treated with chemotherapy. Clin Cancer Res 22(7):1603–1610CrossRef
30.
Zurück zum Zitat Park S, Ha S, Kwon HW et al (2017) Prospective evaluation of changes in tumor size and tumor metabolism in patients with advanced gastric cancer undergoing chemotherapy: association and clinical implication. J Nucl Med 58(6):899–904CrossRef Park S, Ha S, Kwon HW et al (2017) Prospective evaluation of changes in tumor size and tumor metabolism in patients with advanced gastric cancer undergoing chemotherapy: association and clinical implication. J Nucl Med 58(6):899–904CrossRef
31.
Zurück zum Zitat Schneider PM, Eshmuminov D, Rordorf T et al (2018) 18FDG-PET-CT identifies histopathological non-responders after neoadjuvant chemotherapy in locally advanced gastric and cardia cancer: cohort study. BMC Cancer 18:548CrossRef Schneider PM, Eshmuminov D, Rordorf T et al (2018) 18FDG-PET-CT identifies histopathological non-responders after neoadjuvant chemotherapy in locally advanced gastric and cardia cancer: cohort study. BMC Cancer 18:548CrossRef
32.
Zurück zum Zitat Mandard AM, Dalibard F, Mandard JC et al (1994) Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma: clinicopathologic correlations. Cancer 73(11):2680–2686CrossRef Mandard AM, Dalibard F, Mandard JC et al (1994) Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma: clinicopathologic correlations. Cancer 73(11):2680–2686CrossRef
33.
Zurück zum Zitat Borggreve AS, Mook S, Verheij M et al (2018) Preoperative image-guided identification of response to neoadjuvant chemoradiotherapy in esophageal cancer (PRIDE): a multicenter observational study. BMC Cancer 18(1):1006CrossRef Borggreve AS, Mook S, Verheij M et al (2018) Preoperative image-guided identification of response to neoadjuvant chemoradiotherapy in esophageal cancer (PRIDE): a multicenter observational study. BMC Cancer 18(1):1006CrossRef
34.
Zurück zum Zitat Kwee RM, Kwee TC (2014) Role of imaging in predicting response to neoadjuvant chemotherapy in gastric cancer. World J Gastroenterol 20(7):1650–1656CrossRef Kwee RM, Kwee TC (2014) Role of imaging in predicting response to neoadjuvant chemotherapy in gastric cancer. World J Gastroenterol 20(7):1650–1656CrossRef
35.
Zurück zum Zitat Pak KH, Yun M, Cheong JH, Hyung WJ, Choi SH, Noh SH (2011) Clinical implication of FDG-PET in advanced gastric cancer with signet ring cell histology. J Surg Oncol 104(6):566–570 Pak KH, Yun M, Cheong JH, Hyung WJ, Choi SH, Noh SH (2011) Clinical implication of FDG-PET in advanced gastric cancer with signet ring cell histology. J Surg Oncol 104(6):566–570
36.
Zurück zum Zitat Park JC, Lee J-H, Cheoi K et al (2012) Predictive value of pretreatment metabolic activity measured by fluorodeoxyglucose positron emission tomography in patients with metastatic advanced gastric cancer: the maximal SUV of the stomach is a prognostic factor. Eur J Nucl Med Mol Imaging 39(7):1107–1116CrossRef Park JC, Lee J-H, Cheoi K et al (2012) Predictive value of pretreatment metabolic activity measured by fluorodeoxyglucose positron emission tomography in patients with metastatic advanced gastric cancer: the maximal SUV of the stomach is a prognostic factor. Eur J Nucl Med Mol Imaging 39(7):1107–1116CrossRef
37.
Zurück zum Zitat Lee JW, Lee SM, Lee M-S, Shin HC (2012) Role of 18F-FDG PET/CT in the prediction of gastric cancer recurrence after curative surgical resection. Eur J Nucl Med Mol Imaging 39(9):1425–1434CrossRef Lee JW, Lee SM, Lee M-S, Shin HC (2012) Role of 18F-FDG PET/CT in the prediction of gastric cancer recurrence after curative surgical resection. Eur J Nucl Med Mol Imaging 39(9):1425–1434CrossRef
38.
Zurück zum Zitat Kim J, Lim ST, Na CJ et al (2014) Pretreatment F-18 FDG PET/CT parameters to evaluate progression-free survival in gastric cancer. Nucl Med Mol Imaging 48(1):33–40CrossRef Kim J, Lim ST, Na CJ et al (2014) Pretreatment F-18 FDG PET/CT parameters to evaluate progression-free survival in gastric cancer. Nucl Med Mol Imaging 48(1):33–40CrossRef
39.
Zurück zum Zitat Grabinska K, Pelak M, Wydmanski J, Tukiendorf A, d’Amico A (2015) Prognostic value and clinical correlations of 18-fluorodeoxyglucose metabolism quantifiers in gastric cancer. World J Gastroenterol 21(19):5901–5909 Grabinska K, Pelak M, Wydmanski J, Tukiendorf A, d’Amico A (2015) Prognostic value and clinical correlations of 18-fluorodeoxyglucose metabolism quantifiers in gastric cancer. World J Gastroenterol 21(19):5901–5909
40.
Zurück zum Zitat Na SJ, o JH, Park JM et al (2016) Prognostic value of metabolic parameters on preoperative 18F-fluorodeoxyglucose positron emission tomography/computed tomography in patients with stage III gastric cancer. Oncotarget 7(39) Na SJ, o JH, Park JM et al (2016) Prognostic value of metabolic parameters on preoperative 18F-fluorodeoxyglucose positron emission tomography/computed tomography in patients with stage III gastric cancer. Oncotarget 7(39)
41.
Zurück zum Zitat Lee S, Seo HJ, Kim S, Eo JS, Oh SC (2017) Prognostic significance of interim 18 F-fluorodeoxyglucose positron emission tomography-computed tomography volumetric parameters in metastatic or recurrent gastric cancer. Asia Pac J Clin Oncol:1–8 Lee S, Seo HJ, Kim S, Eo JS, Oh SC (2017) Prognostic significance of interim 18 F-fluorodeoxyglucose positron emission tomography-computed tomography volumetric parameters in metastatic or recurrent gastric cancer. Asia Pac J Clin Oncol:1–8
43.
Zurück zum Zitat Gillies RJ, Kinahan PE, Hricak H (2016) Radiomics: images are more than pictures, they are data. Radiology 278(2):563–577CrossRef Gillies RJ, Kinahan PE, Hricak H (2016) Radiomics: images are more than pictures, they are data. Radiology 278(2):563–577CrossRef
44.
Zurück zum Zitat Cook GJR, Azad G, Owczarczyk K, Siddique M, Goh V (2018) Challenges and promises of PET radiomics. Int J Radiat Oncol Biol Phys 102(4):1083–1089CrossRef Cook GJR, Azad G, Owczarczyk K, Siddique M, Goh V (2018) Challenges and promises of PET radiomics. Int J Radiat Oncol Biol Phys 102(4):1083–1089CrossRef
45.
Zurück zum Zitat Lovinfosse P, Visvikis D, Hustinx R, Hatt M (2018) FDG PET radiomics: a review of the methodological aspects. Clin Transl Imaging 6:379–391CrossRef Lovinfosse P, Visvikis D, Hustinx R, Hatt M (2018) FDG PET radiomics: a review of the methodological aspects. Clin Transl Imaging 6:379–391CrossRef
47.
Zurück zum Zitat Jiang Y, Yuan Q, Lv W et al (2018) Radiomic signature of 18F fluorodeoxyglucose PET/CT for prediction of gastric cancer survival and chemotherapeutic benefits. Theranostics 8(21):5915–5928CrossRef Jiang Y, Yuan Q, Lv W et al (2018) Radiomic signature of 18F fluorodeoxyglucose PET/CT for prediction of gastric cancer survival and chemotherapeutic benefits. Theranostics 8(21):5915–5928CrossRef
48.
Zurück zum Zitat Rosenkrantz AB, Mendiratta-Lala M, Bartholmai BJ et al (2015) Clinical utility of quantitative imaging. Acad Radiol 22(1):33–49CrossRef Rosenkrantz AB, Mendiratta-Lala M, Bartholmai BJ et al (2015) Clinical utility of quantitative imaging. Acad Radiol 22(1):33–49CrossRef
Metadaten
Titel
Gastric cancer and image-derived quantitative parameters: Part 2—a critical review of DCE-MRI and 18F-FDG PET/CT findings
verfasst von
Lei Tang
Xue-Juan Wang
Hideo Baba
Francesco Giganti
Publikationsdatum
07.08.2019
Verlag
Springer Berlin Heidelberg
Erschienen in
European Radiology / Ausgabe 1/2020
Print ISSN: 0938-7994
Elektronische ISSN: 1432-1084
DOI
https://doi.org/10.1007/s00330-019-06370-x

Weitere Artikel der Ausgabe 1/2020

European Radiology 1/2020 Zur Ausgabe

Mammakarzinom: Brustdichte beeinflusst rezidivfreies Überleben

26.05.2024 Mammakarzinom Nachrichten

Frauen, die zum Zeitpunkt der Brustkrebsdiagnose eine hohe mammografische Brustdichte aufweisen, haben ein erhöhtes Risiko für ein baldiges Rezidiv, legen neue Daten nahe.

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Klinikreform soll zehntausende Menschenleben retten

15.05.2024 Klinik aktuell Nachrichten

Gesundheitsminister Lauterbach hat die vom Bundeskabinett beschlossene Klinikreform verteidigt. Kritik an den Plänen kommt vom Marburger Bund. Und in den Ländern wird über den Gang zum Vermittlungsausschuss spekuliert.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Radiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.