s11547-023-01625-6

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

La radiologia medica (2023) 128:509–519

https://doi.org/10.1007/s11547-023-01625-6

ABDOMINAL RADIOLOGY

Multi‑modal analysis for accurate prediction of preoperative stage


and indications of optimal treatment in gastric cancer
Shangqing Liu1 · Weiqi Liang2 · Pinyu Huang1 · Dianjie Chen2 · Qinglie He2 · Zhenyuan Ning1 · Yu Zhang1 ·
Wei Xiong3 · Jiang Yu2 · Tao Chen2,4

Received: 14 November 2022 / Accepted: 27 March 2023 / Published online: 28 April 2023
© Italian Society of Medical Radiology 2023

Abstract
Background Accurate preoperative clinical staging of gastric cancer helps determine therapeutic strategies. However, no
multi-category grading models for gastric cancer have been established. This study aimed to develop multi-modal (CT/EHRs)
artificial intelligence (AI) models for predicting tumor stages and optimal treatment indication based on preoperative CT
images and electronic health records (EHRs) in patients with gastric cancer.
Methods This retrospective study enrolled 602 patients with a pathological diagnosis of gastric cancer from Nanfang
hospital retrospectively and divided them into training (n = 452) and validation sets (n = 150). A total of 1326 features were
extracted of which 1316 radiomic features were extracted from the 3D CT images and 10 clinical parameters were obtained
from electronic health records (EHRs). Four multi-layer perceptrons (MLPs) whose input was the combination of radiomic
features and clinical parameters were automatically learned with the neural architecture search (NAS) strategy.
Results Two two-layer MLPs identified by NAS approach were employed to predict the stage of the tumor showed greater
discrimination with the average ACC value of 0.646 for five T stages, 0.838 for four N stages than traditional methods with
ACC of 0.543 (P value = 0.034) and 0.468 (P value = 0.021), respectively. Furthermore, our models reported high prediction
accuracy for the indication of endoscopic resection and the preoperative neoadjuvant chemotherapy with the AUC value of
0.771 and 0.661, respectively.
Conclusions Our multi-modal (CT/EHRs) artificial intelligence models generated with the NAS approach have high
accuracy for tumor stage prediction and optimal treatment regimen and timing, which could facilitate radiologists and
gastroenterologists to improve diagnosis and treatment efficiency.

Keywords Gastric cancer · Artificial intelligence · Multi-modal · Neural architecture search

Abbreviations
EGC Early gastric cancer
EHRs Electronic health records
Shangqing Liu and Weiqi Liang share co-first authorship.
LAGC​ Locally advanced gastric cancer
* Tao Chen AUC​ Area under the receiver operating characteristic
drchentao@163.com curve
1 CA125 Cancer antigen 125
School of Biomedical Engineering, Southern Medical
University, Guangzhou 510515, China CA19-9 Carbohydrate antigen 19-9
2 CEA Carcinoembryonic antigen
Department of General Surgery and Guangdong Provincial
Key Laboratory of Precision Medicine for Gastrointestinal CNN Convolutional Neural Network
Tumor, Nanfang Hospital, The First School of Clinical DL Deep learning
Medicine, Southern Medical University, Guangzhou 510515, MLP Multi-layer perceptron
Guangdong, China NAS Neural architecture search
3
Department of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou 510515, China
4
Department of Gastrointestinal and Hernia Surgery, Ganzhou
Hospital‑Nanfang Hospital, Southern Medical University,
Ganzhou 341000, Jiangxi, China

13
Vol.:(0123456789)
510 La radiologia medica (2023) 128:509–519

Background data can be used to identify patterns for some diseases


and provide useful information for clinicians in their
Gastric cancer (GC) is one of the most common malignant practice. These motivate us to apply machine learning
neoplasms worldwide and causes nearly 800,000 techniques into gastric cancer diagnosis and treatment
deaths per year [1]. TNM staging system is one of the with the combination of CT visual data and EHRs clinical
most important criteria for clinical decision-making information.
and prognostic evaluation of GC, which includes the However, analyzing CT images by radiomics has some
evaluation of the tumor invasion depth (T stage), the technical limitations, including hand-crafted feature
number of regional lymph node metastasis (N stage), and engineering and multi-step pipelines [13]. Recently, DL
the appearance of distant metastasis (M stage) [2]. The methods are becoming increasingly popular for medical
eighth edition of the TNM staging system firstly proposed image analysis because of their impressive classification
the clinical tumor staging method (cTNM) to standardize performance [14]. Yet selecting a model from a wide
the preoperative management of GC patients, and the range of deep network architecture proposed or designed
specific criteria for staging also refer to the pathological is complex and often requires human prior, with a good
diagnosis of the tumor. Early studies have shown that knowledge of the field and the data. Here we propose to
T-stage and N-stage are independent prognostic factors adopt the neural architecture search (NAS) approach for
for survival in GC patients, and the risk of lymph node deep learning model construction. NAS is a framework that
metastasis is higher with the increase in T-stage [3]. automates the process of designing and optimizing neural
Early gastric cancer (EGC) and locally advanced gastric networks for researchers and has recently been actively
cancer (LAGC) are two key time points in the treatment studied in the field of 3D medical image segmentation [15,
of gastric cancer. Anatomically, EGC is defined as the 16].
tumor confined to the mucosa or submucosa, independent This study aimed to develop and optimize a multi-
of lymph node metastasis; LAGC is defined as the tumor modal (CT/EHRs) artificial intelligence model based on
extends beyond the anatomical border of the stomach both preoperative radiomics and clinical information that
without distant metastasis or invasion of the serosa layer. automatically predict the tumor stage of GC patients and
According to statistics, the 5-year survival rate of early help to identify optimal treatment regimen and timing. The
gastric cancer can reach 90%, which is much higher than comparison between the diagnostic results obtained using
the overall survival rate of gastric cancer (25–30%) [4]. our NAS-refined model and those obtained by experienced
At present, the technique of endoscopic gastrectomy for radiologists showed the superiority of rapid and precise
early gastric cancer is rather well-established with smaller diagnosis.
trauma and quicker postoperative recovery than surgical
operation [5]. Despite the poor outcomes, several clinical
trials in recent years have demonstrated that neoadjuvant Methods
chemotherapy significantly reduces the disease staging
and improves radical resection, disease-free survival, and Study population
overall survival rate for LAGC patients [6]. Therefore, the
precise preoperative diagnosis of tumor staging is of great Patients were enrolled based on the following inclusion
significance to improve the survival rate and quality of life criteria: (1) pathologically diagnosed as GC; (2) CT
for GC patients. carried out less than 2 weeks before surgery; (3) complete
Computed tomography (CT) imaging is used widely in clinical and pathological data were available. Patients were
preoperative assessment and clinically staging of gastric excluded based on the following criteria: (1) neoadjuvant
cancer as the CT of the abdomen contains essential visual therapy (chemotherapy, radiotherapy, chemoradiotherapy,
data for the characterization of gastric tissue patterns immunotherapy, molecular targeted therapy, etc.); (2)
associated with gastric cancer [7]. Radiomics can previous abdominal malignancies or inflammatory diseases;
automatically provide many quantitative image features (3) present with unresectable primary tumor lesion or
from medical images, which tend to be hard for naked distant metastases (liver, peritoneal, etc.) during surgical
eyes to recognize [8]. Radiomics-based analysis of GC exploration (4) difficult to segment the tumor because of
CT images has shown good performance on classification, unsatisfactory gastric distention; (5) artifacts on CT images
predicting tumor recurrence and overall survival [9–12]. seriously deteriorating the observation of tumor infiltration
Meanwhile, electronic health records (EHRs) systems depth. A schematic flow chart for all enrolled patients is
collect patient’s health information including diagnosis, shown in Fig. 1.
laboratory tests and medication. Therefore, EHRs A cumulative total of 602 GC patients were enrolled in
our center between January 2005 and December 2015 for

13
La radiologia medica (2023) 128:509–519 511

was ≥ 1 cm, it was considered metastatic lymph nodes. When


the two physicians disagreed on the diagnosis, a third senior
radiologist was consulted to resolve the issue.

Image acquisition and ROI segmentation

After a non-contrast CT scan (Scanner: SIEMENS


64-MDCT or GE Healthcare, Hino) with a thickness of
2·0 mm, a dynamic contrast-enhanced scan was performed,
with 90–100 ml iodine contrast medium (Ultravist 370,
Bayer Schering Pharma, Germany) injected intravenously
at a rate of 3.0–3.5 ml/s. The other acquisition parameters
are as follows: tube voltage, 120 kV; tube current, 160–300
mAs; tube rotation time, 0.5–0.8 s; detector collimation,
8 × 2.5 or 64 × 0.625 mm; field of view, 350 × 350 mm.
Arterial phase image of contrast t-enhanced abdominal
CT with manual region of interest (ROI) was selected for
analysis.
The ITK-SNAP Software (version 3.8.0, http://w ​ ww.i​ tksn​
ap.​org/) was utilized to segment regions of interest (ROIs)
of each tumor on CT images, which were delineated with
the whole data blindly by two radiologists with 12 (reader
1, W.X.) and 7 years (reader 2, Z.C.) of experience in the
Fig. 1  The flowchart of patients enrollment interpretation of abdominal CT (ICC value = 0.93). Before
delineation, grey-level standardization was applied to reduce
the grey-level differences caused by the imaging procedure.
the subsequent analysis (Fig. 2). EHRs were obtained from
All voxels within the ROI were extracted for analysis.
the inpatient records system and the radiological data were
collected from the picture archiving and communication
system (PACS) of our institution as a single-center Radiomic feature extraction
dataset. The retrospective study was approved by the local
ethics committee of our hospital, and the requirement for We extracted visual features in three categories: first-
patient informed consent was waived (Approval Number: order statistics, shape-based features, and texture features,
NFEC-2016-132). including grey level co-occurrence matrix (GLCM) features,
grey level size zone matrix (GLSZM) features, grey level
Reference standard for gastric cancer stage run length matrix (GLRLM) features, neighboring grey
tone difference matrix (NGTDM) features and grey level
The pathologic staging of tumor was based on the 8th edition dependence matrix (GLDM) features. Each feature was
of AJCC/UICC tumor staging system. The clinical staging extracted from the patient’s volume of interest, stacked
of tumor T staging were described as follows: (1) cT1: by the corresponding ROIs delineated slice-by-slice. An
focal thickening (with or without internal enhancement) open-source python package named Pyradiomics (https://​
of the gastric wall, and low-density striations in the basal github.​com/​AIM-​Harva​rd/​pyrad​iomics) was used to extract
part of the lesions near the submucosa; (2) cT2 stage: the radiomics features.
wall of stomach was thickened obviously with the loss or We also applied feature extraction on wavelet and
destruction of low-density striations, but the margin of Laplacian of Gaussian (LoG) filtered images in addition
serosa on the outside of the focus was clear and the fat space to the original images. The wavelet filtering yields
around the stomach was clearly visible; (3) cT3 stage: the eight decompositions per level, including all possible
wall of stomach was thickened obviously at the focus, (4) combinations of applying a high or a low pass filter in each
cT4: the lesion breaks through the serosa and encroaches of the three dimensions. The LoG filter enhances the edge
on the surrounding tissues or adjacent organs [17]. The by emphasizing areas of grey level change. Sigma value in
clinical N staging criteria of the lesions were mainly based the LoG filter defines how coarse the emphasized texture
on the density, enhancement, and size of lymph nodes should be. We extracted features from LoG filtered images
on CT images [18]. When the diameter of lymph nodes with sigma value equals 1.0, 2.0, 3.0, 4.0, 5.0, respectively.

13
512 La radiologia medica (2023) 128:509–519

Fig. 2  The pipeline of model development and model application. connected neural network was developed using the neural architecture
A Collection of clinical data. Digital data of patients were extracted search strategy. D Model evaluation. The classification abilities
from electronic health records (EHRs) and three-dimensional (3D) of our models were assessed by the ROC and classification metrics
segmentation of tumors in CT images were performed. B Radiomic such as accuracy, sensitivity and specificity. During the model
feature extraction and clinical characteristics preprocessing. application process, the new patient information was processed by
Radiomics features, including intensity, shape, and texture features, the constructed model and got the patient-level tumor stage and the
were extracted from ROIs. C Model construction. Radiomic indication of optimal treatment decisions
features and clinical characteristics fused. Finally, a two-layer fully

In total, we extracted 1316 features from each patient. the pipeline of image processing, model development,
Features were divided into three groups: (1) First-order and model application. We use NAS approach to develop
statistics (n = 252); (2) shape (n = 14); (3) texture based four MLP (multi-layer perceptron) models for four tasks:
(n = 1050), including GLCM (n = 336), GLSZM (n = 224), (1) prediction of T-stage; (2) prediction of N-stage; (3)
GLRLM (n = 224), GLDM (n = 196) and NGTDM (n = 70). prediction of the indication for endoscopic procedure; (4)
prediction of the indication for neoadjuvant chemotherapy.
Clinical features extraction We applied NAS using the python package of Optuna, an
open-source hyperparameter optimization framework to
Clinical information collected included age, gender, tumor automate hyperparameter search [19]. Details of model
location, T stage, N stage, degree of differentiation, and development and settings are described in Appendix E1.
tumor markers such as CEA, CA19-9, CA72-4, CA12-5
from EHRs. In total, we extracted 10 clinical features from Model evaluation
each patient. All the values were categorized using integers.
The area under the receiver operating characteristic curve
Model algorithms and development (AUC) was used as the performance measure. Accuracy,
sensitivity, and specificity were calculated too. We
After extracting the clinical and visual features from each calculated their macro average for evaluation. The true
patient, we concatenated these two features and got the positive definition of a specific category is accurately
final multi-modal input which could make the model learn predicted as the category, and the true negative definition is
effectively with more diverse features. Figure 2 presents accurately predicted as one of the remaining four categories.

13
La radiologia medica (2023) 128:509–519 513

Youden Index was calculated to determine the best cutoff Table 1  Patient characteristics in the training and validation cohorts
value to predict the outcomes. Training cohort Validation cohort P value
(N = 452) (N = 150)
Statistical analysis
Age (year) 56.0 ± 11.5 54.5 ± 10.8 0.155
Gender
Statistical analyses were performed using python software
Male 298 (65.9%) 105 (70.0%) 0.413
(version 3.8.3, https://w
​ ww.p​ ython.o​ rg/) with Scipy package
Female 154 (34.1%) 45 (30.0%)
(version 1.5.1, https://​www.​scipy.​org/) or SPSS software
Tumor location
(version 24.0; SPSS Inc., Chicago, IL). Differences between
Up 93 (20.6%) 30 (20.0%) 0.324
cohorts were assessed using t-tests or Mann–Whitney U
Medium 93 (20.6%) 23 (15.3%)
tests for continuous variables; chi-squared tests or Fisher’s
Low 266 (58.8%) 97 (64.7%)
exact test were applied for categorical variables. Receiver
Tumor grade
operating characteristic (ROC) curves were used to display
High 22 (4.9%) 7 (4.7%) 0.78
and evaluate model performance.
Medium 75 (16.6%) 30 (20.0%)
Low 350 (77.4%) 112 (74.7%)
Undifferentiated 5 (1.1%) 1 (0.7%)
Results
cT stage
T1a 16 (3.5%) 3 (2.0%) 0.776
Baseline information of participants
T1b 39 (8.6%) 13 (8.7%)
T2 57 (12.6%) 15 (10.0%)
The 602 patient cohort from the center was divided into a
T3 81 (17.9%) 26 (17.3%)
training cohort (N = 452) and a validation cohort (N = 150).
T4a 245 (54.2%) 86 (57.3%)
Detailed clinicopathological baseline data and comparison
T4b 14 (3.1%) 7 (4.7%)
results are shown in Table 1. The training cohort consisted of
cN stage
298 males and 154 females (mean age 56.0 ± 11.5 years, age
N0 233 (51.5%) 76 (50.7%) 0.656
range 22–83 years), and the validation cohort consisted of
N1 106 (23.5%) 33 (22.0%)
105 males and 45 females (mean age 54.5 ± 10.8 years, age
N2 82 (18.1%) 26 (17.3%)
range 22–79 years). There were no significant differences
N3 31 (6.9%) 15 (10.0%)
in tumor location (longitudinal), tumor differentiation,
cM stage
preoperative CA 19–9 level, and CA 125 level, and
M0 441 (97.6%) 148 (98.7%) 0.632
significant differences in preoperative CEA level and CA
M1 11 (2.4%) 2 (1.3%)
72–4 between these two cohorts.
cTNM
I 94 (20.8%) 23 (15.3%) 0.059
Prediction of tumor stage and feature importance
II A 10 (2.2%) 7 (4.7%)
analysis
II B 125 (27.7%) 34 (22.7%)
III 195 (43.1%) 72 (48.0%)
Based on the Yonden Index, we determined the model
IV A 17 (3.8%) 12 (8.0%)
threshold for each prediction task. The overall accuracy for
IV B 11 (2.4%) 2 (1.3%)
predicting T-stage in the training and validation cohort were
pT stage
0.803 and 0.646, respectively, and the macro-average AUC
T1a 33 (7.3%) 11 (7.3%) 0.912
values were 0.892 and 0.716 (Table E2-3, Fig. 3A, B). In
T1b 51 (11.3%) 13 (8.7%)
the prediction model of lymph node metastasis, the overall
T2 53 (11.7%) 15 (10.0%)
accuracy in the training and validation cohort were 0.669
T3 43 (9.5%) 17 (11.3%)
and 0.838. respectively, and the macro-average AUC values
T4a 234 (51.8%) 81 (54.0%)
were 0.712 and 0.698 (Table E4-5, Fig. 3C, D).
T4b 38 (8.4%) 13 (8.7%)
To better understand the landscape of clinical parameters
pN stage
and radiomic features in the prediction model, we took
N0 184 (40.7%) 49 (32.7%) 0.294
the CNN model as a dark box and got the weight of each
N1 68 (15.0%) 27 (18.0%)
input by permutation technique. In the prediction task of
N2 81 (17.9%) 27 (18.0%)
T-stage, original image derived dependence variance (DV)
N3a 67 (14.8%) 31 (20.7%)
of GLDM applying LoG filter with Gaussian kernel size
N3b 52 (11.5%) 16 (10.7%)
of 4 (log-4_gldm_DependenceVariance) weighted the most
important in the forecasting system (Fig. 4A). The top three

13
514 La radiologia medica (2023) 128:509–519

Table 1  (continued) higher log-4 _gldm_DependenceVariance values tend to


Training cohort Validation cohort P value be more advanced and aggressive (Fig. 5A). For N stage,
(N = 452) (N = 150) the correlation analysis revealed that Original_Shape_
maximum_2D_Diameter was positively correlative with
pM stage
lymph node metastasis (rs = 0.334, P < 0.001) (Fig. 5B).
M0 411 (90.9%) 137 (91.3%) 1.000
M1 41 (9.1%) 13 (8.7%)
Prediction model of optimal treatment regimen
pTNM
and timing based on tumor stage
IA 76 (16.8%) 20 (13.3%) 0.503
IB 34 (7.5%) 12 (8.0%)
Based on the above results, we intended to further
II A 30 (6.6%) 10 (6.7%)
construct predictive models for identifying potential benefit
II B 69 (15.3%) 18 (12.0%)
populations for preoperative neoadjuvant and endoscopic
III A 100 (22.1%) 36 (24.0%)
therapy according to the tumor stage prediction, thereby
III B 56 (12.4%) 29 (19.3%)
avoiding overtreatment by surgery. According to the
III C 46 (10.2%) 12 (8.0%)
literature, the preoperative staging of gastric cancer patients
IV 41 (9.1%) 13 (8.7%)
with indications for endoscopic resection (IER) should be
CEA (ng/mL)
T1aN0M0, we therefore screened patients with pT1aN0M0
≥  5 213 (47.1%) 56 (37.3%) 0.046*
staging (44/602, 7.3%) from the entire cohort and labeled
<5 239 (52.9%) 94 (62.7%)
them as predictors; we also manually selected patients with
CA 19–9 (U/mL)
indication for preoperative neoadjuvant chemotherapy (INC,
≥ 37 211 (46.7%) 56 (37.3%) 0.057
pT1-2N + M0 or pT3/4NxM0, 464/602, 77.1%) to further
< 37 241 (53.3%) 94 (62.7%)
our research. The results showed that the accuracy of IER in
CA 72-4 (U/mL)
the training and validation cohort were 1.000 and 0.718, and
≥6 162 (35.8%) 39 (26.0%) 0.034*
the AUC value were 1.000 and 0.771 (Table E4, Fig. 6). In
<6 290 (64.2%) 111 (74.0%)
the task of predicting INC, the accuracy of the training and
CA 125 (U/mL)
validation cohort were 0.720 and 0.713, and the AUC value
≥ 35 7 (1.5%) 4 (2.7%) 0.593
were 0.687 and 0.661, respectively. (Table E4, Fig. 6). In the
< 35 445 (98.5%) 146 (97.3%)
two models, the most important feature for predicting IER
*P < 0.05 and INC were wavelet-LHH_ngtdm_Strength and original_
glrlm_RunLengthNonUniformity, respectively (Fig. 6).

factors that played critical roles in the predicting process


were: log-4_gldm_DependenceVariance, original_glcm_ Discussion
Inverse-Variance, log-5_gldm_SmallDependenceHigh-
GrayLevelEmphasis. In the prediction task of N-stage, The aim of this study was to develop and validate a multi-
original image derived maximum 2D Diameter row ranked modal (CT/EHRs) artificial intelligence model for predicting
highest above other features (Fig. 4B), and the following the tumor stages and the optimal treatment regimen and
two factors are log-3_glrlm_LongRunEmphasis, and timing of GCs patients with NAS approach. To determine
wavelet_LLLfirstorder_TotalEnergy. Notably, CA 19–9 its performance for grading tumor stages, we reported the
exhibited great contribution to both prediction models as macro-average AUC of 0.716 for five T stages and 0.698 for
the only clinical feature that appears in the top 1% of all four N stages, respectively. Meanwhile, we also reported
input variables. the macro-average ACC of 0.646 for five T stages and
0.838 for four N stages, respectively, which showed greater
Correlation between the important features discrimination than traditional methods with ACC of 0.543
and tumor stages (P value = 0.034) and 0.468 (P value = 0.021), respectively.
Meanwhile, we reported the AUC of 0.771 for the indication
From the top five important features mentioned above, for endoscopic resection and 0.661 for the indication for
we managed to further discover the relationship between preoperative neoadjuvant chemotherapy, respectively.
input features and T-stage as well as N-stage. Using During CT screening procedures, these models may assist
Spearman’s correlation analysis, positive correlations doctors in predicting the histology of ambiguous lesions and
were found between log- 4 _gldm_DependenceVariance, determining diagnostic or therapeutic strategies.
Original_glcm_InverseVarianceand and T-stage (rs = 0.277, Although guidelines for the treatment of GC vary from
P < 0.001; rs = 0.157, P < 0.001). In general, tumors with country to country, overall treatment principles are based

13
La radiologia medica (2023) 128:509–519 515

Fig. 3  ROC curves of the prediction model. A infiltration depth model in the training cohort; D lymph node metastasis prediction
prediction model in the training cohort; B infiltration depth prediction model in the validation cohort
model in the validation cohort; C lymph node metastasis prediction

on the surgeon's accurate preoperative clinical staging information to classify T-stage in binary categories for GC
and risk stratification. Moreover, early identification of such as T2 vs T3/T4 and T1/T2/T3 vs T4 successfully with
LAGC patients before surgery can improve the response the AUC values of 0.818–0.825 and 0.813–0.840 in the
rate of neoadjuvant chemotherapy and provide further testing cohort [11, 22]; Chen et al. and Wang et al. applied
treatment for patients who lost the chance of surgery. the DL method to diagnostic analysis such as recurrence
According to the literature, the accuracy rate of Radiologists prediction and metastasis identification [23, 24]. However,
subjectively determining the depth of invasion and lymph most studies adopted a single examination method, and did
node metastasis of GC based on CT images are about 70%, not integrate multi-modal examination methods.
which is unsatisfactory [20, 21]. In previous studies, Wang In this study, we combined enhanced CT visual data with
et al. and Meng et al. used traditional radiomics and clinical the EHRs data to construct multi-modal models to focus on

13
516 La radiologia medica (2023) 128:509–519

Fig. 4  Top 10 important features for gastric cancer A infiltration depth prediction model; B lymph node metastasis prediction model

Fig. 5  Spearman’s correlation plots of tumor infiltration depth (A) and lymph node metastasis (B) with their top important feature, respectively

the prediction of specific stages of GC into five T categories growth, which from the view of graphic processing is
and four N categories as well as the identification to recive the relationship between a voxel and neighboring voxels.
endoscopic resection and neoadjuvant chemotherapy Consequently, the histogram-like function of GLDM
preoperatively to simulate real-world clinical decision- were able to illustrate the color transitions between
making. Given a limited number of training samples, adjacent pixels with better performance. As for clinical
training multiple sophisticated deep learning models are parameters, CA19-9 gives a robust contribution to the
very challenging due to the choice of the hyperparameters classification task. Carbohydrate antigen 19-9 has been
and construction of the networks architecture. Fortunately, widely used for assisting diagnosis and treatment efficacy
we can use the NAS approach adaptively to design the best evaluation in digestive tract malignancies like pancreatic
architecture and hyperparameters to tackle these challenges. cancer or cholangiocarcinoma [25, 26]. The association
From the feature importance analysis, we can see that of elevated CA19-9 levels with gastric carcinoma has
the top 10 important features have both radiomic features been presented in a case report and other studies [27,
from CT images and clinical parameters from EHRs. This 28]. Elevated CA19-9 levels have been significantly
means that the multi-modal (CT/EHRs) information is correlated with lymph node metastasis, vascular invasion,
more helpful for gastric cancer diagnosis and treatment and liver metastasis. Our results indicated that CA19-9
decisions compared with single modal CT images or aids in T-stage and N-stage differentiation, consistent with
EHRs. In terms of visual information, radiomic features previous researches [29, 30].
extracted from the original image filtered by wavelet are We found that the model performed poorly in
less critical than LoG and the original image. The most intermediate classes such as T3 or T4a (Table E2–E3). An
significant feature in this study originated from the GLDM explanation for this phenomenon was that the category of
method of measuring variance independence size in CT T-stage generally depends on the location and volume of
images. We considered this finding to be relevant to the the primary tumor lesion. The difference between the sizes
interpretation of images. T-stage describes the expansion of some intermediate tumors may be subtle. Secondly, there
of the lesion border within the gastric wall during tumor existed a severe class imbalance in our data, consistent with

13
La radiologia medica (2023) 128:509–519 517

Fig. 6  ROC curves of the optimal clinical decision model and Top 10 important features of each model, respectively. A ROC curve of the IER
model; B ROC curve of the INC model; C important features in the IER model; D important features in the INC model

the real-world clinical practice in our country that the GC need to be considered. Firstly, this was a retrospective study
patients graded T1/T2 is relatively fewer than T3/T4 [31]. conducted at a single center with inter-class imbalance.
The key strengths of this study include the use of multi- Secondly, there is still room for the accuracy and overall
modal (CT/EHRs) information with the combination of performance of our model to improve. Thirdly, the prediction
visual features and clinical features, and the use of NAS of the indications of neoadjuvant and endoscopic therapy were
approach to develop p the artificial intelligence model which only a stimulation of clinicians in clinical decision-making
alleviates the challenge for the choice of hyperparameters and based on clinical tumor stage prior to surgery, thus it can only
networks architecture during training. In detail, we built two provide reference rather than medical instructions. Fourthly,
multi-category prediction model which is nearly consistent a multicenter study with a larger dataset is needed to further
with pathological findings using radiomics features from CT validate our models’ reproducibility and generalization ability
images and clinical parameters. This will help doctors save a in future studies.
certain amount of time spend on diagnosis, so as to gain more
treatment time for patients. To our knowledge, our method is
the first work combining the radiomics and NAS approach to
develop the AI models, which alleviates the challenge for the
choice of hyperparameters and network architecture during
training. However, there are also some potential limitations

13
518 La radiologia medica (2023) 128:509–519

Conclusions 4. Oliveira C, Pinheiro H, Figueiredo J, Seruca R, Carneiro F


(2015) Familial gastric cancer: genetic susceptibility, pathology,
and implications for management. Lancet Oncol 16(2):e60-70
To sum up, this AI-based staging model incorporating 5. Roy S, Kanda M, Nomura S et al (2022) Diagnostic efficacy of
high-dimensional radiomics features and clinical circular RNAs as noninvasive, liquid biopsy biomarkers for early
information possess the potential to aid preoperative detection of gastric cancer. Mol Cancer 21(1):42
6. Schernberg A, Rivin Del Campo E, Rousseau B et al (2018)
clinical tumor stage classification, filter early or locally Adjuvant chemoradiation for gastric carcinoma: state of the art
advanced GC cases and make reasonable recommendations and perspectives. Clin Transl Radiat Oncol 10:13–22
in the clinical decision-making process. 7. Barros RH, Penachim TJ, Martins DL, Andreollo NA, Caserta NM
(2015) Multidetector computed tomography in the preoperative
Supplementary Information The online version contains staging of gastric adenocarcinoma. Radiol Bras 48(2):74–80
supplementary material available at https:// ​ d oi. ​ o rg/ ​ 1 0. ​ 1 007/​ 8. Gillies RJ, Kinahan PE, Hricak H (2016) Radiomics: images are
s11547-​023-​01625-6. more than pictures, they are data. Radiology 278(2):563–577
9. Giganti F, Antunes S, Salerno A et al (2017) Gastric cancer:
Acknowledgements Not applicable. texture analysis from multidetector computed tomography
as a potential preoperative prognostic biomarker. Eur Radiol
Author contributions All authors have had access to the data and 27(5):1831–1839
all drafts of the manuscript. SL, WL contributed equally as co-first 10. Chen X, Yang Z, Yang J et al (2020) Radiomics analysis of
authors. Specific contributions are as follows: SL: conceptualization, contrast-enhanced CT predicts lymphovascular invasion and
methodology, software, investigation, writing - review & editing; WL: disease outcome in gastric cancer: a preliminary study. Cancer
conceptualization, resources, data curation, visualization, writing - Imaging 20:1–12
original draft; PH: formal analysis, writing - original draft; DC: data 11. Meng L, Dong D, Chen X et al (2021) 2D and 3D CT radiomic
curation, formal analysis; QH: validation, visualization; ZN: writing features performance comparison in characterization of gastric
- review & editing, project administration; YZ: project administration, cancer: a multi-center study. IEEE J Biomed Health Inform
supervision; WX: writing - review & editing, supervision; JY: project 25(3):755–763
administration, supervision; TC: project administration, supervision, 12. Li J, Dong D, Fang M et al (2020) Dual-energy CT–based deep
funding acquisition. All authors read and approved the final manuscript. learning radiomics can improve lymph node metastasis risk
prediction for gastric cancer. Eur Radiol 30(4):2324–2333
Funding This project was supported by Guangdong Provincial Key 13. Koçak B, Durmaz E, Ateş E, Kılıçkesmez Ö (2019) Radiomics
Laboratory of Precision Medicine for Gastrointestinal Cancer (No. with artificial intelligence: a practical guide for beginners. Diagn
2020B121201004), Guangdong Natural Science Foundation General Interv Radiol 25(6):485–495
Project (2019A1515011520) and Guangdong Natural Science 14. Lee JH, Ha EJ (2020) Application of deep learning to the
Foundation Outstanding Youth Project (2021B1515020055). diagnosis of cervical lymph node metastasis from thyroid cancer
with CT: external validation and clinical utility for resident
Data availability All data generated or analyzed during this study are training. Eur Radiol 30(6):3066–3072
included either in this paper or in the additional information. 15. Zoph B, Le QV (2016) Neural architecture search with
reinforcement learning. arXiv preprint arXiv:​1611.​01578.
Declarations 16. Bae W, Lee S, Lee Y, Park B, Chung M, Jung K-H (2019)
Resource optimized neural architecture search for 3D medical
Coonflict of interest The authors declare that they have no competing image segmentation. In: International conference on medical
interests. image computing and computer-assisted intervention. Springer,
pp 228–236
Ethics approval and consent to participate The Institutional Review 17. Kim JW, Shin SS, Heo SH et al (2012) Diagnostic performance
Board of Nanfang Hospital of Southern Medical University approved of 64-section CT using CT gastrography in preoperative T staging
this study and waived the need for informed consent from patients of gastric cancer according to 7th edition of AJCC cancer staging
(Approval Number: NFEC-2016-132). manual. Eur Radiol 22(3):654–662
18. Hallinan JT, Venkatesh SK, Peter L, Makmur A, Yong WP, So
Consent for publication Not applicable. JB (2014) CT volumetry for gastric carcinoma: association with
TNM stage. Eur Radiol 24(12):3105–3114
19. Akiba T, Sano S, Yanase T, Ohta T, Koyama M (2019) Optuna:
a next-generation hyperparameter optimization framework. In:
Proceedings of the 25th ACM SIGKDD international conference
on knowledge discovery & data mining, pp 2623–2631
References 20. Seevaratnam R, Cardoso R, McGregor C et al (2012) How useful
is preoperative imaging for tumor, node, metastasis (TNM)
1. Sung H, Ferlay J, Siegel RL (2021) Global cancer statistics staging of gastric cancer? A meta-analysis Gastric Cancer
2020: GLOBOCAN estimates of incidence and mortality 15(Suppl 1):S3-18
worldwide for 36 cancers in 185 countries. CA Cancer J Clin 21. Choi JI, Joo I, Lee JM (2014) State-of-the-art preoperative staging
71(3):209–249 of gastric cancer by MDCT and magnetic resonance imaging.
2. Amin M, Edge S, Greene F (2017) AJCC cancer staging manual. World J Gastroenterol 20(16):4546–4557
Springer, Berlin 22. Wang Y, Liu W, Yu Y et al (2020) Prediction of the depth of tumor
3. Xu YY, Huang BJ, Sun Z, Lu C, Liu YP (2007) Risk factors for invasion in gastric cancer: potential role of CT radiomics. Acad
lymph node metastasis and evaluation of reasonable surgery for Radiol 27(8):1077–1084
early gastric cancer. World J Gastroenterol 13(38):5133–5138 23. Chen T, Liu S, Li Y et al (2019) Developed and validated a
prognostic nomogram for recurrence-free survival after complete

13
La radiologia medica (2023) 128:509–519 519

surgical resection of local primary gastrointestinal stromal tumors early detection and monitoring progression of gastric cancer: a
based on deep learning. EBioMedicine 39:272–279 multiphase study. JAMA Surg 155(7):572–579
24. Wang X, Chen Y, Gao Y et al (2021) Predicting gastric cancer 30. Song XH, Liu K, Yang SJ et al (2020) Prognostic value of changes
outcome from resected lymph node histopathology images using in preoperative and postoperative serum CA19-9 levels in gastric
deep learning. Nat Commun 12(1):1637 cancer. Front Oncol 10:1432
25. Carpelan-Holmström M, Louhimo J, Stenman UH, Alfthan H, 31. Tsendsuren T, Jun SM, Mian XH (2006) Usefulness of endoscopic
Haglund C (2002) CEA, CA 19-9 and CA 72-4 improve the ultrasonography in preoperative TNM staging of gastric cancer.
diagnostic accuracy in gastrointestinal cancers. Anticancer Res World J Gastroenterol 12(1):43–47
22(4):2311–2316
26. Ballehaninna UK, Chamberlain RS (2012) The clinical utility Publisher's Note Springer Nature remains neutral with regard to
of serum CA 19-9 in the diagnosis, prognosis and management jurisdictional claims in published maps and institutional affiliations.
of pancreatic adenocarcinoma: an evidence based appraisal. J
Gastrointest Oncol 3(2):105–119 Springer Nature or its licensor (e.g. a society or other partner) holds
27. Feng F, Tian Y, Xu G et al (2017) Diagnostic and prognostic value exclusive rights to this article under a publishing agreement with the
of CEA, CA19-9, AFP and CA125 for early gastric cancer. BMC author(s) or other rightsholder(s); author self-archiving of the accepted
Cancer 17(1):737 manuscript version of this article is solely governed by the terms of
28. Jo JC, Ryu MH, Koo DH et al (2013) Serum CA 19–9 as a such publishing agreement and applicable law.
prognostic factor in patients with metastatic gastric cancer. Asia
Pac J Clin Oncol 9(4):324–330
29. Guo X, Lv X, Ru Y et al (2020) Circulating exosomal gastric
cancer-associated long noncoding RNA1 as a biomarker for

13

You might also like