这是用户在 2024-5-7 14:16 为 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10315777/ 保存的双语快照页面,由 沉浸式翻译 提供双语支持。了解如何保存?
Skip to main content
跳到主要内容
U.S. flag

An official website of the United States government

Access keys 访问密钥 NCBI Homepage NCBI主页 MyNCBI Homepage MyNCBI主页 Main Content 事由 Main Navigation 主导航
2023 Jun; 12(12): 13111–13122.
癌症医学 2023 年 6 月;12(12): 13111–13122.
Published online 2023 May 3. doi: 10.1002/cam4.5994
2023 年 5 月 3 日在线发布。doi: 10.1002/cam4.5994
PMCID: PMC10315777 PMCID:PMC10315777
PMID: 37132269 PMID:37132269

Establishment and validation of a prognostic nomogram for postoperative patients with gastric cardia adenocarcinoma: A study based on the Surveillance, Epidemiology, and End Results database and a Chinese cohort
胃贲门腺癌术后患者预后列线图的建立和验证:一项基于监测、流行病学和最终结果数据库和中国队列的研究

Lei Wang, 1 Jingjing Ge, 1 Liwen Feng, 1 Zehua Wang, 1 Wenjia Wang, 1 Huiqiong Han,corresponding author 1 and Yanru Qincorresponding author 1
王磊, 1 葛晶晶, 冯 1 立文, 1 王泽华, 1 王文佳, 1 韩慧琼, corresponding author 1 corresponding author 1 彦茹

Associated Data 相关数据

Data Availability Statement
数据可用性声明

Abstract 抽象

Background 背景

Gastric cardia adenocarcinoma (GCA) is a highly fatal form of cancer in humans. The aim of this study was to extract clinicopathological data of postoperative patients with GCA from the Surveillance, Epidemiology, and End Results database, analyze prognostic risk factors, and build a nomogram.
胃贲门腺癌 (GCA) 是一种高度致命的人类癌症。本研究的目的是从监测、流行病学和最终结果数据库中提取 GCA 术后患者的临床病理学数据,分析预后危险因素,并构建列线图。

Methods 方法

In this study, the clinical information of 1448 patients with GCA who underwent radical surgery and were diagnosed between 2010 and 2015 was extracted from the SEER database. The patients were then randomly divided into training (n = 1013) and internal validation (n = 435) cohorts at a 7:3 ratio. The study also included an external validation cohort (n = 218) from a Chinese hospital. The study used the Cox and LASSO models to pinpoint the independent risk factors linked to GCA. The prognostic model was constructed according to the results of the multivariate regression analysis. To assess the predictive accuracy of the nomogram, four methods were used: C‐index, calibration curve, time‐dependent ROC curve, and DCA curve. Kaplan–Meier survival curves were also generated to illustrate the differences in cancer‐specific survival (CSS) between the groups.
本研究从SEER数据库中提取了2010-2015年间接受根治性手术诊断的1448例GCA患者的临床信息。然后将患者以 7:3 的比例随机分为训练组 (n = 1013) 和内部验证组 (n = 435)。该研究还包括来自中国医院的外部验证队列(n = 218)。该研究使用Cox和LASSO模型来确定与GCA相关的独立风险因素。根据多因素回归分析结果构建预后模型。为了评估列线图的预测准确性,使用了四种方法:C指数、校准曲线、瞬态ROC曲线和DCA曲线。还生成了Kaplan-Meier生存曲线,以说明两组之间癌症特异性生存期(CSS)的差异。

Results 结果

The results of the multivariate Cox regression analysis showed that age, grade, race, marital status, T stage, and log odds of positive lymph nodes (LODDS) were independently associated with cancer‐specific survival in the training cohort. Both the C‐index and AUC values depicted in the nomogram were greater than 0.71. The calibration curve revealed that the nomogram's CSS prediction was consistent with the actual outcomes. The decision curve analysis suggested moderately positive net benefits. Based on the nomogram risk score, significant differences in survival between the high‐ and low‐risk groups were observed.
多因素Cox回归分析结果显示,在训练队列中,年龄、年级、种族、婚姻状况、T分期和阳性淋巴结对数(LODDS)的对数几率与癌症特异性生存率独立相关。列线图中描述的 C 指数和 AUC 值均大于 0.71。校准曲线显示列线图的CSS预测与实际结果一致。决策曲线分析表明净收益为中等正。根据列线图风险评分,观察到高危组和低危组之间的生存率存在显著差异。

Conclusions 结论

Race, age, marital status, differentiation grade, T stage, and LODDS are independent predictors of CSS in patients with GCA after radical surgery. Our predictive nomogram constructed based on these variables demonstrated good predictive ability.
种族、年龄、婚姻状况、分化等级、T 分期和 LODDS 是根治性手术后 GCA 患者 CSS 的独立预测因子。基于这些变量构建的预测列线图表现出良好的预测能力。

Keywords: gastric cardia adenocarcinoma, LODDS, nomogram, prognosis, SEER database
关键词:胃贲门腺癌,LODDS,列线图,预后,SEER数据库

Short abstract 简短摘要

The nomogram for postoperative patients with GCA we constructed has good predictive power and can help clinicians accurately assess patient prognosis and identify high‐risk patients to develop more personalized treatment plans.
我们构建的GCA术后患者列线图具有良好的预测能力,可以帮助临床医生准确评估患者预后,识别高危患者,制定更加个性化的治疗方案。

1. INTRODUCTION 1. 引言

Gastric cardia adenocarcinoma (GCA) is a commonly diagnosed malignant tumor of the digestive tract. Although the overall incidence of gastric cancer (GC) has recently declined globally, the incidence of GCA is still increasing., GCA originates at the independent zone of the esophagogastric junction (EGJ), which differs from GC that originates at other sites in terms of pathophysiology. GCA remains a challenging disease, and radical surgery is currently the only available management method. Early diagnosis and timely surgery can have a positive impact on the prognosis of patients with GCA. In addition, despite advances in treatment, the prognosis for patients with GCA remains suboptimal due to factors such as tumor recurrence., However, the introduction of personalized treatment has directed renewed attention to the prognostic factors that impact patients with cancer. Therefore, it is significant to analyze the prognostic risk factors and construct a survival prediction model for patients with GCA after radical surgery.
胃贲门腺癌(GCA)是一种常见诊断的消化道恶性肿瘤。 尽管最近全球胃癌 (GC) 的总体发病率有所下降,但 GCA 的发病率仍在上升。 , GCA 起源于食管胃交界处 (EGJ) 的独立区,在病理生理学方面与起源于其他部位的 GC 不同。 GCA仍然是一种具有挑战性的疾病,根治性手术是目前唯一可用的治疗方法。早期诊断和及时手术可以对GCA患者的预后产生积极影响。此外,尽管治疗取得了进展,但由于肿瘤复发等因素,GCA 患者的预后仍然不理想。 , 然而,个性化治疗的引入重新引起了人们对影响癌症患者的预后因素的关注。因此,分析根治性手术后GCA患者的预后危险因素并构建生存预测模型具有重要意义。

The TNM staging system is currently the primary method used to evaluate patient prognosis and guide clinical treatment approaches. However, relying solely on the TNM staging system has obvious limitations, as it lacks important variables such as age, differentiation grade, and other influential factors. Therefore, it is not suitable for individualized analysis., Multiple studies have demonstrated that the log odds of positive lymph nodes (LODDS) are superior to the N stage alone in determining lymph node metastasis and providing accurate prognostic assessments for patients with cancer., In line with this, a nomogram is a graphical calculation instrument based on statistical models that can predict the likely outcomes. Each variable is assigned a score based on its degree of risk, and the final sum of all scores corresponds to the predicted survival probability. Prior research has demonstrated that nomograms have a better prediction performance compared to that of TNM staging, leading to the construction of various nomograms to predict prognosis in different types of cancer., , ,
TNM分期系统是目前用于评估患者预后和指导临床治疗方法的主要方法。 然而,仅依靠TNM分期系统具有明显的局限性,因为它缺乏年龄、分化等级和其他影响因素等重要变量。因此,它不适合进行个体化分析。 , 多项研究表明,在确定淋巴结转移和为癌症患者提供准确的预后评估方面,阳性淋巴结 (LODDS) 的对数几率优于单独的 N 期。 , 与此一致,列线图是一种基于统计模型的图形计算工具,可以预测可能的结果。 每个变量都根据其风险程度分配一个分数,所有分数的最终总和对应于预测的生存概率。先前的研究表明,与TNM分期相比,列线图具有更好的预测性能,导致构建各种列线图来预测不同类型癌症的预后。 , , ,

The Surveillance, Epidemiology, and End Results (SEER) database covers over 28% of the population of the United States and includes information on patient demographics, stage of diagnosis, treatment process, tumor morphology, primary tumor site, vital status follow‐up, and causes of death, thus providing an effective tool for tumor epidemiological research. The objective of this study was to obtain clinical data from the SEER database and use it to analyze the prognostic risk factors in postoperative patients with GCA. The study also aimed to construct a nomogram model that could accurately predict cancer‐specific survival (CSS) in patients with GCA and evaluate the nomogram through internal and external validation.

2. METHODS

2.1. Patient selection

The specific operational process for extracting information from the SEER database is outlined below: (1) Register for a personal account on the SEER database official website and install the SEER*Stat software; (2) Extract clinical data of patients based on the inclusion and exclusion criteria determined in this study; (3) Export the extracted data as a spreadsheet and proceed to the next step of organizing and analyzing the data.

The data for patients with GCA between 2010 and 2015 were downloaded from the SEER*Stat 8.4.0 software using a private ID (13914‐Nov2021), based on the 2021 release of the SEER database. According to the SEER codes, patients with adenocarcinoma (ICD‐O‐3 codes: 8140–8145,8147, 8210, 8211, 8214, 8220, 8221, 8230, 8231, 8255, 8260–8263, 8310, 8480, 8481, 8490, 8510, 8560, 8562, 8570–8576) and tumors located in the cardia (site code: C16.0) were included. All patients underwent radical surgery (surgery encode 30–80); however, those with stage IV GCA were excluded due to the controversial nature of their operation. In addition, patients with missing information on relevant variables were excluded from this study. The detailed screening process and selection criteria are shown in Figure 1. The patients included in this study were randomly divided into training (70%) and internal validation (30%) groups.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g004.jpg

Flowchart for the selection of the patients.

In addition to the above data, the data of patients with GCA after surgery at the First Affiliated Hospital of Zhengzhou University between 2012 and 2018 were collected for validation. Patients underwent thorough preoperative assessments, including physical exams, medical history recording, hematological testing, and imaging. Furthermore, tumor metastasis was evaluated using various imaging techniques. The study was approved by the hospital ethics committee, and patient records were kept confidential in accordance with ethical standards.

2.2. Variable collection

According to the instructions provided by the National Cancer Institute, the code data extracted from the SEER database was translated. The external validation cohort data was obtained through the hospital medical record system. The variables collected in this study were mainly divided into three categories: patient‐related variables, disease‐related variables, and follow‐up information. (1) Patient‐related variables: race, sex, age at diagnosis, and marital status. (2) Disease‐related variables: histological type, differentiation grade, 7th edition AJCC clinical stage (TNM), tumor size, examined lymph nodes (ELN), positive lymph nodes (PLN), and LODDS, which is calculated as LODDS = log[(PLN + 0.5)/(ELN‐PLN + 0.5)]. (3) Follow‐up information: survival status, cause of death, and survival time.

Each variable was categorized as follows: marital status, unmarried or married; sex, male or female; histological type, adenocarcinoma or signet ring cell carcinoma; differentiation grade, G1–2 (well to moderately differentiated) or G3–4 (poorly differentiated and undifferentiated); and the 7th edition AJCC clinical stage, which divided T stage into T1, T2, T3, and T4 and N stage into N0, N1, N2, and N3. The X‐tile software was used to determine the optimal cutoff values for the three continuous variables of age, tumor size, and LODDS. Age was classified as <65, 65–71, and >71 years; tumor size was classified as <2.6, 2.6–4.8, and <4.8 cm; LODDS was classified as LODDS1 (< −1.20), LODDS2 (−1.20 to −0.60), and LODDS3 (< −0.60).

2.3. Statistical analysis

SPSS (26.0) and R (4.2.2) software were used for statistical analysis and graph plotting. The X‐tile software determined the optimal cutoff value, and continuous variables were transformed into categorical variables accordingly. Categorical variables were compared between groups using the Fisher's exact test or chi‐square test. Univariate Cox analysis was conducted for each variable, and variables with statistical significance (p < 0.05) were included in the LASSO equation for feature selection. In addition, the multivariate Cox analysis was performed to identify independent predictive factors. A two‐tailed p < 0.05 was considered statistically significant.

Based on the multivariate Cox regression analysis, a nomogram was constructed to predict patient CSS using the rms and survival packages in R software. The model's reliability was validated by internal and external validation cohorts. The model's discrimination was evaluated using the concordance index (C‐index), with a C‐index greater than 0.71 indicating excellent discrimination. The calibration curve measured the degree of closeness between the predicted and actual risk, with a closer curve indicating better predictive results. The time‐dependent receiver operating characteristic curve (ROC curve) evaluated the model's accuracy, with an area under the curve (AUC) greater than 0.71 indicating good predictive ability. Decision curve analysis (DCA) evaluated the model's clinical utility and quantified the net benefits at different threshold probabilities. Patients were divided into high‐risk and low‐risk groups based on the median risk score from the column chart. Kaplan–Meier analysis was used to plot the survival curves of high‐risk and low‐risk groups for predicting CSS, and the log‐rank test was used for survival analysis.

3. RESULTS

3.1. Patient characteristics

Data of 1448 patients with GCA who underwent curative surgery were extracted from the SEER database, and these patients were randomly assigned to a training set (1013 patients) and an internal validation set (435 patients) at a ratio of 7:3. In the training cohort, the median follow‐up time was 41 months, with 3‐year and 5‐year CSS rates of 57.7% and 47.3%, respectively. In the internal validation cohort, the median follow‐up time was 37 months, with 3‐year and 5‐year CSS rates of 55.5% and 43.9%, respectively. In the training cohort, there were 818 male (80.8%) and 195 female (19.2%) patients. Of the total sample, 86.2% (n = 873) were White, 4.6% (n = 47) were Black, and 9.2% (n = 93) belonged to other racial categories. Overall, 555 patients aged <65 years (54.8%), 241 aged between 65 and 71 years (23.8%), and 217 aged >71 years (21.4%). In the internal validation cohort, there were 366 male (84.1%) and 69 female (15.9%) patients. Of the total sample, 88.1% (n = 383) were White, 4.8% (n = 21) were Black, and 7.1% (n = 31) belonged to other racial categories. In this cohort, 230 patients aged <65 years (52.9%), 97 aged between 65 and 71 years (22.3%), and 108 aged >71 years (24.8%). The overall grouping of the training and internal validation cohorts was consistent with simple random grouping.

Retrospective clinical data of 218 patients with GCA who underwent curative surgery at a Chinese hospital were collected, and these patients were assigned to an external validation cohort according to the same inclusion and exclusion criteria, and the nomogram model was validated with external data. In the external validation cohort, the median follow‐up time was 31 months, with 3‐year and 5‐year CSS rates of 69.0% and 58.6%, respectively. There were 174 male (79.8%) and 44 female (20.2%) patients in the external validation cohort. In this cohort, 136 patients aged <65 years (62.4%), 57 aged between 65 and 71 years (26.1%), and 25 aged >71 years (11.5%). The basic information is presented in Table 1.

TABLE 1

Clinicopathological characteristics of the postoperative patients with stage I–III GCA.

VariableNO. (%)
Training cohortInternal validation cohortExternal validation cohort
Sex
Female195 (19.2)69 (15.9)44 (20.2)
Male818 (80.8)366 (84.1)174 (79.8)
Race
White873 (86.2)383 (88.1)0 (0)
Black47 (4.6)21 (4.8)0 (0)
Other93 (9.2)31 (7.1)218 (100)
Age, years
<65555 (54.8)230 (52.9)136 (62.4)
65–71241 (23.8)97 (22.3)57 (26.1)
>71217 (21.4)108 (24.8)25 (11.5)
Marital status
Unmarried294 (29.0)129 (29.7)7 (3.2)
Married719 (71.0)306 (70.3)211 (96.8)
Histological type
Adenocarcinoma907 (89.5)400 (92.0)206 (94.5)
Signet ring carcinoma106 (10.5)35 (8.0)12 (5.5)
Tumor grade
Well442 (43.6)189 (43.4)59 (27.1)
Poor571 (56.4)246 (56.6)159 (72.9)
TNM stage
I189 (18.7)78 (17.9)14 (6.4)
II275 (27.1)114 (26.2)68 (31.2)
III549 (54.2)243 (55.9)136 (62.4)
T stage
T1200 (19.8)80 (18.4)8 (3.7)
T2143 (14.1)69 (15.9)20 (9.2)
T3609 (60.1)260 (59.8)37 (16.9)
T461 (6.0)26 (5.9)153 (70.2)
N stage
N0382 (37.7)157 (36.1)67 (30.7)
N1346 (34.2)136 (31.3)47 (21.6)
N2170 (16.8)86 (19.8)51 (23.4)
N3115 (11.3)56 (12.8)53 (24.3)
Size, cm
<2.6310 (30.6)144 (33.1)29 (13.3)
2.6–4.8364 (35.9)156 (35.9)102 (46.8)
>4.8339 (33.5)135 (31.0)87 (39.9)
LODDS
LODDS1456 (45.0)194 (44.6)67 (30.7)
LODDS2288 (28.4)125 (28.7)46 (21.1)
LODDS3269 (26.6)116 (26.7)105 (48.2)

3.2. Independent prognostic factors

The univariate Cox regression analysis showed that race, age, marital status, grade, T stage, N stage, tumor size, and LODDS score were related to CSS. To avoid overfitting, LASSO regression was performed using those eight variables. All eight variables were included in the model, as their coefficients were non‐zero (Figure 2). The variables that showed significance in the univariate and LASSO regression analyses were included in the multivariate analysis. In the multifactorial Cox analysis, age (65–71 and >71 years), grade (poor), T stage (T1–3), and LODDS (LODDS2 and LODDS2) were independent prognostic risk factors in the patients with GCA at stages I–III. Moreover, race (other) and marital status (married) were protective factors that showed better patient prognosis. Table 2 displays the outcomes of both univariate and multivariate Cox analyses conducted on the training set.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g007.jpg

LASSO coefficients of eight features (A) and the selection of tuning parameter (λ) for the LASSO model (B).

TABLE 2

Univariate and multivariate cox regression analyses of the prognostic factors for CSS.

VariablesUnivariate analysisMultivariate analysis
HR (95% CI) p‐valueHR (95% CI) p‐value
Sex
Female1
Male0.989 (0.796–1.229)0.922
Race
White11
Black1.121 (0.765–1.641)0.5580.910 (0.616–1.346)0.638
Other0.621 (0.440–0.875) 0.007 0.621 (0.439–0.877) 0.007
Age, years
<6511
65–711.240 (1.006–1.527) 0.043 1.348 (1.091–1.666) 0.006
>711.445 (1.168–1.788) <0.001 1.472 (1.184–1.830) <0.001
Marital status
Unmarried11
Married0.787 (0.654–0.946) 0.011 0.742 (0.614–0.897) 0.002
Histological type
Adenocarcinoma11
Signet ring carcinoma1.096 (0.832–1.443)0.514
Tumor grade
Well11
Poor1.809 (1.513–2.163) <0.001 1.405 (1.168–1.689) <0.001
T stage
T111
T22.044 (1.402–2.980) <0.001 1.613 (1.091–2.383) 0.016
T33.569 (2.553–4.800) <0.001 2.304 (1.649–3.218) <0.001
T46.029 (4.025–9.032) <0.001 2.868 (1.826–4.505) <0.001
N stage
N011
N12.183 (1.735–2.746) <0.001 1.004 (0.758–1.329)0.979
N23.742 (2.907–4.816) <0.001 1.152 (0.820–1.620)0.415
N34.885 (3.703–6.443) <0.001 0.983 (0.661–1.461)0.931
Size, cm
<2.611
2.6–4.81.736 (1.374–2.194) <0.001 1.101 (0.855–1.420)0.456
>4.82.445 (1.943–3.076) <0.001 1.282 (0.993–1.656)0.056
LODDS
LODDS111
LODDS22.420 (1.935–3.028) <0.001 2.266 (1.740–2.950) <0.001
LODDS35.222 (4.198–6.496) <0.001 3.869 (2.843–5.266) <0.001

The bold font represents p < 0.05, indicating a statistically significant variable.

3.3. Establishment of the nomogram

Based on the results of the multivariate Cox regression analysis, a nomogram was established and is presented in Figure 3. The C‐index values for the training, internal validation, and external validation sets were 0.737 (95% confidence interval [CI], 0.717–0.757), 0.733 (95% CI, 0.702–0.764), and 0.746 (95% CI, 0.689–0.803), respectively. The calibration curves in Figure 4 show that the predicted results are consistent with the observed results. As shown in Figure 5, the tdROCs showed that the AUC values of the nomogram in the 3‐ and 5‐year groups were 0.802 (95% CI, 0.774–0.830) and 0.818 (95% CI, 0.789–0.847) in the training set, 0.796 (95% CI, 0.753–0.839) and 0.804 (95% CI, 0.755–0.853) in the internal validation set, and 0.805 (95% CI, 0.732–0.877) and 0.836 (95% CI, 0.746–0.925) in the external validation set, respectively. In comparison, the AUC values of TNM stage in the 3‐ and 5‐year groups were 0.692 (95% CI, 0.663–0.722) and 0.717 (95% CI, 0.683–0.751) in the training set, 0.715 (95% CI, 0.671–0.758) and 0.744 (95% CI, 0.693–0.796) in the internal validation set, and 0.712 (95% CI, 0.646–0.778) and 0.758 (95% CI, 0.655–0.862) in the external validation set, respectively. The nomogram had superior predictive ability compared to that of the TNM stage alone (p < 0.05). Furthermore, the DCA curves in Figure 6 show that the nomogram for cancer‐CSS provides significant clinical benefits. In summary, the predictive model based on the aforementioned factors had a strong predictive power for CSS with high accuracy and clinical utility in patients who underwent radical surgery for GCA. This was supported by the results of various statistical analyses such as the –index, calibration curve, ROC, and DCA.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g006.jpg

Nomogram predicting CSS of the postoperative patients with GCA.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g003.jpg

Calibration curves of the nomogram for predicting 3‐year CSS (A) and 5‐year CSS (B) in the training set; the calibration curves of the nomogram for predicting 3‐year CSS (C) and 5‐year CSS (D) in the internal validation set; and the calibration curves of the nomogram for predicting 3‐year CSS (E) and 5‐year CSS (F) in the external validation set.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g002.jpg

Time‐dependent ROC curves were used to test the predictive power of the 3‐year CSS and 5‐year CSS in the training se (A and D); the internal validation set (B and E); and the external validation set (C and F), respectively.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g008.jpg

DCA of the nomogram and TNM stage 3‐year CSS and 5‐year CSS of the training (A and D), internal (B and E), and external cohorts (C and F), respectively.

3.4. Risk stratification based on the nomogram

The risk score was calculated using the nomogram, and patients were categorized into low‐risk and high‐risk groups based on the median value as the cutoff point. The Kaplan–Meier plot (Figure 7) showed that patients in the low‐risk group had a significantly better prognosis than those in the high‐risk group (p < 0.001). Further analysis (Figure 8) revealed that chemotherapy was beneficial only in the high‐risk group, as identified by our model. This suggests that our model can aid physicians in identifying high‐risk patients who may benefit from chemotherapy, allowing for personalized treatment plans.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g005.jpg

Kaplan–Meier curves for the patients in the low‐ and high‐risk groups based on the risk scores. (A) training cohort; (B) internal validation set; and (C) external validation set.

An external file that holds a picture, illustration, etc.
Object name is CAM4-12-13111-g001.jpg

Effect of chemotherapy on the survival in the total population (A), low‐risk group (B), and high‐risk group (C).

4. DISCUSSION

Currently, mainstream studies consider the EGJ to be a region that is separate from both the esophagus and the stomach. Given that GCA is a type of malignant tumor occurring in the EGJ that has caused the death of millions of individuals, this disorder warrants further studies. Surgical resection is undoubtedly the most important treatment approach for GCA, with a 5‐year survival rate of 43–49%. However, despite advancements in treatment, the survival rates for postoperative patients with GCA remain below desired levels, and many experience recurrence and death each year. While the TNM staging system is widely used as a prognostic assessment tool to guide postoperative treatment protocols, it has limitations in terms of specificity and does not consider important patient factors such as age, sex, and marital status. As a result, the predictive ability of TNM staging may not be sufficiently accurate. In contrast, nomograms are one of the most widely used forecasting tools that can comprehensively consider multiple factors, including clinical pathology and demographic characteristics. This is why our research study focused on developing a nomogram for predicting CSS in postoperative patients with GCA. Previous studies have reported different prognostic models for adenocarcinomas of the EGJ., , However, most of the previously built models lacked external validation, which reduced their applicability. Therefore, we aimed to construct a predictive model for the prognosis of postoperative patients with GCA at stages I–III GCA and enhance the reliability of the model through validation in an external population.

Our study aimed to construct a nomogram that accurately predicts the prognosis of postoperative patients with GCA at stages I–III based on a multivariate analysis. The C‐index and AUC values were both greater than 0.71, indicating favorable discrimination of the nomogram. Compared to the TNM staging system alone, the nomogram model had a higher accuracy, as shown by the AUC value. The DCA curve was used to analyze the clinical benefits of the model, and the results suggested that the nomogram had a high net clinical benefit. Further statistical analyses showed that the nomogram model had more advantages than the TNM staging system. The use of our nomogram model aligns with the current principles of personalized treatment. Finally, the applicability of the model was verified using an external population.

Although chemotherapy is widely used in the treatment of GCA, our study found that it did not have a positive therapeutic effect on patients in this study. However, the CLASSIC trial established the benefits of adjuvant capecitabine and oxaliplatin in patients with GC/AEG who underwent surgery. Based on the nomogram score in this study, we stratified the study population into high‐risk and low‐risk groups. We observed that chemotherapy had a positive therapeutic effect only in the high‐risk group. This indicates that the nomogram‐based identification of high‐risk groups is accurate and can guide clinicians in formulating personalized chemotherapeutic regimens. As for the low‐risk population, the results of our study suggest that chemotherapy may not provide significant therapeutic benefits. Therefore, overtreatment should be avoided to prevent unnecessary adverse effects and improve the patient's quality of life.

In the multifactor analysis, race, age, marital status, histological grade, T stage, and LODDS were determined to be independent prognostic factors. It has been widely reported that older age is a poor prognostic factor in cancer patients., For instance, elderly patients often have functional impairments, malnutrition, and comorbidities that prompt physicians to choose less aggressive treatments or shorten the course of treatment, which in turn affects the treatment outcomes. In contrast, it has been suggested that younger patients may have a better tolerance to the adverse effects of treatment, including myelosuppression after chemotherapy, compared with that in elderly patients. Additionally, in this study, unmarried patients were more prone to anxiety and had greater stress burdens than married patients, thereby reducing their immune capacity and affecting their metabolic balance, resulting in decreased survival., In addition, unmarried patients had poorer compliance with treatment and received the treatment later, which may also be a reason for their poor prognoses.

Zhu et al. observed that White patients had the highest risk of GCA compared with that in the other ethnic groups, which is consistent with our findings. In prior studies, race‐related differences in morbidity and outcomes have been attributed to obesity and unequal incomes., In addition, lymph node metastasis has been found to have a significant impact on postoperative outcomes, including long‐term survival and postoperative adjuvant therapy administration. The results of our study showed that LODDS was a more significant prognostic factor than the conventional N stage in assessing postoperative risk factors for GCA. This is consistent with results of previous studies, suggesting that more attention should be paid to the LODDS of postoperative patients rather than solely focusing on the number of positive lymph nodes., , , Additionally, tumors with poor differentiation are generally more aggressive and have a higher likelihood of recurrence and distant metastasis, requiring close monitoring of this patient group.

Compared with the study by Guo et al., our study included more variables. Additionally, we conducted an external validation, which enhanced the reliability of our model. To the best of our knowledge, this is the first nomogram to predict the survival of postoperative patients with GCA with both internal and external population validation. However, our study has some limitations. First, important clinical information was lacking from the SEER database. Smoking, alcohol consumption, body mass index, diet, performance, and family history are of great significance in the prognostic evaluation of malignant tumors. In addition, this study did not include molecular or genetic information that is used in routine clinical treatments, such as EGFR mutations and Her‐2 expression. Additionally, the SEER database lacks information on specific regimens of chemoradiotherapy, targeted therapy, and immunotherapy, which could have affected the results of our study. Another limitation of our study is that it was retrospective in nature, and thus, a prospective study is necessary to further validate our findings. This will be the focus of our subsequent studies.

5. CONCLUSIONS

In conclusion, we utilized clinical data from the SEER database to identify factors associated with survival in postoperative patients with GCA at stages I–III. Subsequently, we developed a nomogram that accurately predicted CSS in patients with GCA who underwent radical surgery. Our findings indicate that the nomogram outperforms TNM staging in terms of predictive power and may provide greater clinical benefits for patients with GCA after radical surgery.

AUTHOR CONTRIBUTIONS

Lei Wang: Conceptualization (lead); methodology (lead); writing – original draft (lead). Jingjing Ge: Conceptualization (equal); methodology (equal); writing – original draft (equal). liwen feng: Data curation (supporting); visualization (supporting). Zehua Wang: Data curation (supporting); visualization (supporting). Wenjia Wang: Data curation (supporting); visualization (supporting). Huiqiong Han: Conceptualization (equal); writing – review and editing (equal). Yanru Qin: Conceptualization (lead); writing – review and editing (lead).

CONFLICT OF INTEREST STATEMENT

The authors have no conflict of interest to declare.

ETHICS APPROVAL STATEMENT

The First Affiliated Hospital of Zhengzhou University's Medical Ethics Committee examined and approved this study involving human participants (2023‐KY‐0019‐001).

PATIENT CONSENT STATEMENT

As this was a retrospective study, no written informed consent was required.

ACKNOWLEDGMENTS

We would like to thank our families for their constant support and encouragement during this study. Their love and sacrifices have been invaluable to our success. We are deeply grateful for their contributions to our personal and professional growth. Thank you, from the bottom of our hearts. No funding was received for this study.

Notes

Wang L, Ge J, Feng L, et al. Establishment and validation of a prognostic nomogram for postoperative patients with gastric cardia adenocarcinoma: A study based on the Surveillance, Epidemiology, and End Results database and a Chinese cohort. Cancer Med. 2023;12:13111‐13122. doi: 10.1002/cam4.5994 [PMC free article] [PubMed] [CrossRef] []

Lei Wang and Jingjing Ge contributed equally as first authors.

Contributor Information

Huiqiong Han, moc.361@32388qhh.

Yanru Qin, nc.ude.uzz@niqurnay.

DATA AVAILABILITY STATEMENT

The datasets utilized in this study can be accessed upon reasonable request to the corresponding author.

REFERENCES

1. Wang Z, Graham DY, Khan A, et al. Incidence of gastric cancer in the USA during 1999 to 2013: a 50‐state analysis. Int J Epidemiol. 2018;47(3):966‐975. [PMC free article] [PubMed] []
2. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. [PubMed] []
3. Ichihara S, Uedo N, Gotoda T. Considering the esophagogastric junction as a ‘zone’. Dig Endosc. 2017;29(Suppl 2):3‐10. [PubMed] []
4. Liu K, Feng F, Chen XZ, et al. Comparison between gastric and esophageal classification system among adenocarcinomas of esophagogastric junction according to AJCC 8th edition: a retrospective observational study from two high‐volume institutions in China. Gastric Cancer. 2019;22(3):506‐517. [PMC free article] [PubMed] []
5. Guo Q, Peng Y, Yang H, Guo J. Prognostic nomogram for postoperative patients with gastroesophageal junction cancer of No distant metastasis. Front Oncol. 2021;11:643261. [PMC free article] [PubMed] []
6. Amin MB, Greene FL, Edge SB, et al. The eighth edition AJCC cancer staging manual: continuing to build a bridge from a population‐based to a more “personalized” approach to cancer staging. CA Cancer J Clin. 2017;67(2):93‐99. [PubMed] []
7. Zhu X, Pigazzi A, Zell J, Lu Y. Changing disparity of gastric cancer incidence by histological types in US race‐specific populations. Cancer Control. 2020;27(1):1073274820977152. [PMC free article] [PubMed] []
8. Wen D, Wen J, Zou W, et al. Site‐specific variation in familial cancer as suggested by family history, multiple primary cancer, age at onset, and sex ratio associated with upper, middle, and lower third esophageal and gastric cardia carcinoma. Front Oncol. 2020;10:579379. [PMC free article] [PubMed] []
9. Cao ZX, Weng X, Huang JS, Long X. Prognostic value of LODDS in medullary thyroid carcinoma based on competing risk model and propensity score matching analysis. Updat Surg. 2022;74(5):1551‐1562. [PubMed] []
10. Patel KN, Salunke A, Bhatt S, et al. Log ODDS (LODDS) of positive lymph nodes as a predictor of overall survival in squamous cell carcinoma of the penis. J Surg Oncol. 2021;123(8):1836‐1844. [PubMed] []
11. Park SY. Nomogram: an analogue tool to deliver digital knowledge. J Thorac Cardiovasc Surg. 2018;155(4):1793. [PubMed] []
12. Shi L, Wang Z, Wang L, Jia Y, Li J, Qin Y. A prognostic nomogram and heat map to predict survival in stage II/III gastric cancer patients after curative gastrectomy followed by adjuvant chemotherapy. Cancer Manag Res. 2022;14:287‐301. [PMC free article] [PubMed] []
13. Feng LW, Li J, Liang LF, et al. A predictive scoring system based on inflammatory and tumor markers for gastric cancer patients undergoing curative resection. Cancer Manag Res. 2020;12:3937‐3948. [PMC free article] [PubMed] []
14. Liu C, Hu C, Huang J, et al. A prognostic nomogram of colon cancer with liver metastasis: a study of the US SEER database and a Chinese cohort. Front Oncol. 2021;11:591009. [PMC free article] [PubMed] []
15. Wang Q, Wang Y, Wang X, et al. Development and validation of a dynamic survival nomogram for metastatic non‐small cell lung cancer based on the SEER database and an external validation cohort. Transl Lung Cancer Res. 2022;11(8):1678‐1691. [PMC free article] [PubMed] []
16. Miller KD, Nogueira L, Devasia T, et al. Cancer treatment and survivorship statistics. CA Cancer J Clin. 2022;72(5):409‐436. [PubMed] []
17. Chen Y, Zhao XK, Xu RH, et al. Transthoracic, thoracoabdominal, and transabdominal surgical approaches for gastric cardia adenocarcinomas: a survival evaluation based on a cohort of 7103 patients. World J Surg Oncol. 2022;20(1):217. [PMC free article] [PubMed] []
18. D'Journo XB. Clinical implication of the innovations of the 8(th) edition of the TNM classification for esophageal and esophago‐gastric cancer. J Thorac Dis. 2018;10(Suppl 22):S2671‐s81. [PMC free article] [PubMed] []
19. Chen K, Deng X, Yang Z, et al. Sites of distant metastases and the cancer‐specific survival of metastatic Siewert type II esophagogastric junction adenocarcinoma: a population‐based study. Expert Rev Gastroenterol Hepatol. 2020;14(6):491‐497. [PubMed] []
20. Liu F, Zhou R, Jiang F, Liu G, Li K, Zhu G. Proposal of a nomogram for predicting survival in patients with Siewert type II adenocarcinoma of the Esophagogastric junction after preoperative radiation. Ann Surg Oncol. 2019;26(5):1292‐1300. [PMC free article] [PubMed] []
21. Joshi SS, Badgwell BD. Current treatment and recent progress in gastric cancer. CA Cancer J Clin. 2021;71(3):264‐279. [PMC free article] [PubMed] []
22. Al‐Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro‐oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet (London, England). 2019;393(10184):1948‐1957. [PubMed] []
23. Lee DU, Fan GH, Chang K, et al. The clinical impact of advanced age on the postoperative outcomes of patients undergoing gastrectomy for gastric cancer: analysis across US hospitals between 2011‐2017. J Gastric Cancer. 2022;22(3):197‐209. [PMC free article] [PubMed] []
24. Brown LR, Kamarajah SK, Madhavan A, et al. The impact of age on long‐term survival following gastrectomy for gastric cancer. Ann R Coll Surg Engl. 2022;105:269‐277. [PMC free article] [PubMed] []
25. Burdett N, Vincent AD, O'Callaghan M, Kichenadasse G. Competing risks in older patients with cancer: a systematic review of geriatric oncology trials. J Natl Cancer Inst. 2018;110(8):825‐830. [PubMed] []
26. Guidi J, Lucente M, Sonino N, Fava GA. Allostatic load and its impact on health: a systematic review. Psychother Psychosom. 2021;90(1):11‐27. [PubMed] []
27. Lee S, Ma C, Zhang S, et al. Marital status, living arrangement, and cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803 (Alliance). Oncologist. 2022;27(6):e494‐e505. [PMC free article] [PubMed] []
28. Krajc K, Miroševič Š, Sajovic J, et al. Marital status and survival in cancer patients: a systematic review and meta‐analysis. Cancer Med. 2023;12(2):1685‐1708. [PMC free article] [PubMed] []
29. Islami F, DeSantis CE, Jemal A. Incidence trends of esophageal and gastric cancer subtypes by race, ethnicity, and age in the United States, 1997‐2014. Clin Gastroenterol Hepatol. 2019;17(3):429‐439. [PubMed] []
30. Lindblad M, Rodríguez LA, Lagergren J. Body mass, tobacco and alcohol and risk of esophageal, gastric cardia, and gastric non‐cardia adenocarcinoma among men and women in a nested case‐control study. Cancer Causes Control. 2005;16(3):285‐294. [PubMed] []
31. Zheng Y, Lu Z, Shi X, et al. Lymph node ratio is a superior predictor in surgically treated early‐onset pancreatic cancer. Front Oncol. 2022;12:975846. [PMC free article] [PubMed] []
32. Xu J, Cao J, Wang L, et al. Prognostic performance of three lymph node staging schemes for patients with Siewert type II adenocarcinoma of esophagogastric junction. Sci Rep. 2017;7(1):10123. [PMC free article] [PubMed] []
33. Sun Z, Xu Y, Li de M, et al. Log odds of positive lymph nodes: a novel prognostic indicator superior to the number‐based and the ratio‐based N category for gastric cancer patients with R0 resection. Cancer. 2010;116(11):2571‐2580. [PubMed] []
34. Tóth D, Bíró A, Varga Z, Török M, Árkosy P. Comparison of different lymph node staging systems in prognosis of gastric cancer: a bi‐institutional study from Hungary. Chin J Cancer Res. 2017;29(4):323‐332. [PMC free article] [PubMed] []
35. Spolverato G, Ejaz A, Kim Y, et al. Prognostic performance of different lymph node staging systems after curative intent resection for gastric adenocarcinoma. Ann Surg. 2015;262(6):991‐998. [PubMed] []
36. Shi X, Xu L, Ma B, Wang S. Development and validation of a nomogram to predict the prognosis of patients with gastric cardia cancer. Sci Rep. 2020;10(1):14143. [PMC free article] [PubMed] []

Articles from Cancer Medicine are provided here courtesy of Wiley-Blackwell