Skip to main content
Erschienen in: BMC Gastroenterology 1/2023

Open Access 01.12.2023 | Research

Prognosis prediction in esophageal signet-ring-cell carcinoma: a competing risk analysis

verfasst von: Chen Chen, Zehua Wang, Yanru Qin

Erschienen in: BMC Gastroenterology | Ausgabe 1/2023

Abstract

Objective

This study aims to construct and validate a competing risk nomogram model to predict 1-year, 3-year, and 5-year cancer-specific survival (CSS) for patients with esophageal signet-ring-cell carcinoma.

Methods

Patients diagnosed with esophageal signet-ring-cell carcinoma (ESRCC) between 2010 and 2015 were abstracted from the Surveillance, Epidemiology, and End Results (SEER) database. We performed the competing risk model to select significant variables to build a competing risk nomogram, which was used to estimate 1-year, 3-year, and 5-year CSS probability. The C-index, receiver operating characteristic (ROC) curve, calibration plot, Brier score, and decision curve analysis were performed in the internal validation.

Results

A total of 564 patients with esophageal signet-ring-cell carcinoma fulfilled the eligibility criteria. The competing risk nomogram identified 4 prognostic variables, involving the gender, lung metastases, liver metastases, and receiving surgery. The C indexes of nomogram were 0.61, 0.75, and 0.70, respectively for 5-year, 3-year, and 1-year CSS prediction. The calibration plots displayed high consistency. The Brier scores and decision curve analysis respectively favored good prediction ability and clinical utility of the nomogram.

Conclusions

A competing risk nomogram for esophageal signet-ring-cell carcinoma was successfully constructed and internally validated. This model is expected to predict 1-year, 3-year, and 5-year CSS, and help oncologists and pathologists in clinical decision making and health care management for esophageal signet-ring-cell carcinoma patients.
Hinweise
Chen Chen and Zehua Wang contributed equally to this work.

Publisher's Note

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

Introduction

Esophageal cancer (EC) is a common malignancy of digestive system and is reported to have 604100 new cases globally in 2020, with Asia accounting for 79.7% [1]. Although esophageal cancer is often diagnosed as squamous-cell carcinomas, [2, 3] the incidence of adenocarcinoma has increased over the past two decades [4]. Esophageal signet-ring cell carcinoma (ESRCC) is a special subtype of esophageal adenocarcinoma characterized by the abundant accumulation of intracellular mucin, which replaces and compresses the nucleus to the periphery of the cell, thereby forming the classical signet-ring shape [5]. As reported in the western studies, the incidence of ESRCC is estimated to float from 3.5% to 5% of all esophageal malignancies [68].
The current understanding of prognostic implications of ESRCC is limited and somewhat inconsistent. Owing to most SRC carcinoma deriving from stomach or gastroesophageal junction, the prognosis of SRC carcinoma primarily originated from esophagus is not well studied [9]. Compared to usual-type esophageal adenocarcinoma, available literature pointed out that ESRCC patients tend to present with more aggressive feature, less responsive to induction therapy, and have worse overall survival (OS) and disease-free survival (DFS) after surgery, all of which support SRC histology as a predictive marker for poor prognosis [6, 7, 1012].
However, one research from the United Stated claimed that SRC does not necessarily portend a worse prognosis [13]. These discrepancies motivate more efforts to provide an overview of survival analysis and find out potential risk factors affecting long-term prognosis. In addition, large population-based study never specifically spotlighted on the prognosis prediction model for ESRCC. The Surveillance, Epidemiology, and End Results (SEER) database covering around 28% U.S. population provides a relatively large sample-based database for exploring such a rare cancer type [14]. Moreover, there are frequently multiple time-to-event outcomes in clinical follow-up. Death is not only attributed to primary tumor or relapse, but also caused by non-ESRCC-specific reasons, such as toxicity of interventions patients underwent. These different outcomes are in competition and might lead to result bias if only performing Cox proportional hazard model. Therefore, the competing risk analysis is more appropriate when dealing with multiple event outcomes.
To address above issues, this study is to identify risk factors affecting cancer-specific survival (CSS) in patients with ESRCC. Besides, we aim to construct and validate a competing risk nomogram model in SEER database to predict 1-year, 3-year, and 5-year cancer-specific survival.

Materials and methods

Study population

The Surveillance, Epidemiology, and End Results (SEER) Program is one of the largest population-based cancer databases, covering approximately 27.8% of the U.S. population [14]. Because of the de-identified nature of SEER database, local institutional ethics committee and informed consents were not required. All patient data were obtained from the SEER database using SEER*Stat, version 8.3.8. A total of 90544 patients were excluded due to missing or incomplete clinic pathological data. Eligible patients (N = 564) with ESRCC were randomly divided into training set (N = 282) and validation set (N = 282) with a ratio of 1:1. A detailed inclusion algorithm is shown in Fig. 1.
The inclusion criteria were set as follows: 1) Patients with esophageal cancer diagnosed between 2010 and 2015; 2) Histology ICD-O-3 (International Classification of Diseases for Oncology) limited to ESRCC (8490/3); 3) Patients with a defined clinical stage (American Joint Committee on Cancer Staging Manual, the 7th edition) including stage I-IV; 4) Follow-up time ≥ 1 month.

Predictive variates and main outcomes

The study covariates were collected and sorted: age at diagnosis (≤ 65 and > 65), sex (male and female), race (white and others), marital status (divorced, married, single, and widowed), history of malignancy (newly and previously), primary tumor site (15-32 cm, 33–40 cm, and others), grade of differentiation (I-II and III-IV), AJCC stage (I, II, III, and IV), T stage (T1, T2, T3, T4, and TX), N stage (N0, N1, N2, and N3/NX), regional lymph node examined (0, 0–10, and 11–60), positive regional lymph nodes (0, 0–2, 3–35), bone metastases (no and yes), liver metastases (no and yes), lung metastases (no and yes), surgery (no and yes), radiotherapy (no and yes), chemotherapy (no and yes).
Death owing to ESRCC was deemed as CSS. Death due to non-cancer reasons were regarded as other-cause death. Overall survival (OS) was defined as the period from the date of initial diagnosis to the date of death or the last follow-up.

Statistical analysis

All statistical analyses were performed with R software version 4.0.5. A P-value of < 0.05 was considered statistically significant.
Covariates were presented as frequency (percentages) and were compared by chi-square test. Cumulative incidences of death (CID) for both ESRCC and non-ESRCC were assessed by the cumulative incidence function (CIF) model. We used the proportional sub distribution hazard (SH) model to quantify the influences of covariates on ESRCC, which is a semi-parametric model considering competing risks. We used the “cmprsk” R package to perform the competing risk analysis. Stepwise regression was conducted in multivariate competing risk analysis to find out the significant variables. The measures of prognostic value on ESRCC-related mortality were displayed with hazard ratios (HR) and 95% confidence interval (CI). All variates were selected based on the minimum Bayesian Information Criterions (BIC) principle. Finally, the determined factors were used to construct a competing risk nomogram model.
Internal validation of the nomogram model was carried out using the validation cohort. Calibration and discrimination are two important aspects to predict the accuracy of the competing risk model. First, the concordance index (C-index) was used to evaluate the prediction accuracy of the established model, ranging from 0.5 (no discrimination) to 1.0 (best discrimination). The Area Under the Curve (AUC) value was measured to assess discrimination ability at the given time. The AUC value was between 0.5 and 0.6, 0.6 and 0.7, or greater than 0.8, manifesting poor, moderate, or good prediction performance, respectively. Second, calibration curves were used to evaluate the consistency between the “predicted value” and the “actual value”. Third, we assessed the Brier score of the nomogram to make a comprehensive evaluation. Finally, we used decision curve analysis (DCA) to judge clinical utility of competing risk model. In DCA curves, threshold probability (X-axis) is the minimum probability of which further strategy would be warranted, and Y-axis represents net benefit that is calculated by subtracting harms form benefits. There are two control curves, representing all patients were treated with the competing risk nomogram and none, respectively.

Results

Patients characteristics

A total of 564 patients with ESRCC fulfilled the eligibility criteria between 2010 and 2015 from the SEER database and were randomly divided into the training cohort (N = 282) and the validation cohort (N = 282). The demographics and clinicopathological characteristics for two groups are presented in Table 1.
Table 1
Baseline characteristics of patients diagnosed with esophageal signet-ring-cell carcinoma
Variables
Total (n = 564)
Training (n = 282)
Validation (n = 282)
P value
No
%
No
%
No
%
Age at Diagnosis
      
0.063
 Mean ± SD
66.64 ± 10.9
 
65.96 ± 11.09
 
67.31 ± 10.69
 
0.142
 Age <  = 65
261
46
142
50
119
42
 
 Age > 65
303
54
140
50
163
58
 
Sex
      
0.523
 Female
70
12
38
13
32
11
 
 Male
494
88
244
87
250
89
 
Race
      
1
 Others
41
7
20
7
21
7
 
 White
523
93
262
93
261
93
 
Marital Status
      
0.892
 Divorced/Separated
55
10
29
10
26
9
 
 Married
343
61
167
59
176
62
 
 Single/Unmarried
93
16
48
17
45
16
 
 Widowed/Others
73
13
38
13
35
12
 
History of Malignancy
      
1
 Newly
543
96
272
96
271
96
 
 Previously
21
4
10
4
11
4
 
Primary Site
      
0.507
 15-32 cm
42
7
20
7
22
8
 
 33-40 cm
468
83
231
82
237
84
 
 Others
54
10
31
11
23
8
 
Grade
      
0.979
 I-II
25
4
13
5
12
4
 
 III-IV
457
81
228
81
229
81
 
 Others
82
15
41
15
41
15
 
AJCC Stage
      
0.26
 I
67
12
26
9
41
15
 
 II
120
21
60
21
60
21
 
 III
192
34
101
36
91
32
 
 IV
185
33
95
34
90
32
 
Tumor Size
      
0.114
 T1
133
24
56
20
77
27
 
 T2
59
10
26
9
33
12
 
 T3
243
43
127
45
116
41
 
 T4
74
13
44
16
30
11
 
 TX
55
10
29
10
26
9
 
Node Status
      
0.823
 N0
196
35
103
37
93
33
 
 N1
257
46
124
44
133
47
 
 N2
64
11
31
11
33
12
 
 N3/NX
47
8
24
9
23
8
 
Regional Nodes Examined
      
0.344
 0
415
74
204
72
211
75
 
 0–10
52
9
31
11
21
7
 
 11–60
97
17
47
17
50
18
 
Positive Lymph Nodes
      
0.682
 0
485
86
240
85
245
87
 
 0–2
44
8
22
8
22
8
 
 3–35
35
6
20
7
15
5
 
Bone Metastases
      
1
 NO/Unknown
514
91
257
91
257
91
 
 YES
50
9
25
9
25
9
 
Liver Metastases
      
1
 NO/Unknown
516
91
258
91
258
91
 
 YES
48
9
24
9
24
9
 
Lung Metastases
      
0.693
 NO/Unknown
537
95
267
95
270
96
 
 YES
27
5
15
5
12
4
 
Surgery
      
0.855
 NO
391
69
194
69
197
70
 
 YES
173
31
88
31
85
30
 
Radiotherapy
      
0.495
 NO
424
75
208
74
216
77
 
 YES
140
25
74
26
66
23
 
Chemotherapy
      
0.213
 NO
148
26
67
24
81
29
 
 YES
416
74
215
76
201
71
 
Outcomes
      
0.639
 Survival
113
20
52
18
61
22
 
 Cancer-specific death
355
63
181
64
174
62
 
 Other-cause death
96
17
49
17
47
17
 
A majority of ESRCC patients were men in both training set (87%) and validation set (89%, P = 0.523). Approximately 93% patients were white among selected population. More than half of the patients (61%) were married. Most patients with ESRCC were newly diagnosed (96%) versus only 4% previously confirmed. Primary tumor sites were frequently detected at 33–40 cm (83%) from the incisors. The proportion of patients who had grade III-IV (81%) ESRCC was remarkably higher than other differentiation grades. Fewer ESRCC patients presented with distant metastasis, regardless of whether the metastases located in the brain, liver, or lung. In the whole group, a lower proportion of patients received surgery (31% VS. 69%) or radiotherapy (25% VS. 75%), while a higher percentage of patients underwent chemotherapy (74% VS. 26%).
However, there are no statistically significant differences (all p > 0.05) between two cohorts in terms of the age at diagnosis (P = 0.063), sex (P = 0.523), race (P = 1.000), marital status (P = 0.892), history of malignancy (P = 1.000), primary site (P = 0.507), grade (P = 0.979), AJCC stage (P = 0.260), tumor size (P = 0.114), node status (P = 0.823), regional nodes examined (P = 0.344), positive lymph nodes (P = 0.682), bone metastasis (P = 1.000), liver metastasis (P = 1.000), lung metastasis (P = 0.693), surgery (P = 0.855), radiotherapy (P = 0.495), chemotherapy (P = 0.213). Therefore, the baseline characteristics were well balanced between two groups.

Cumulative incidence of ESRCC

We classified the time-to-event outcomes into two endpoints: cancer-specific death and other-cause death. For all included patients (N = 564), there were 355 ESRCC-specific deaths, 96 other causes of deaths, and 113 alive patients during the follow-up. The 5-year cumulative incidence plots depicting cancer-caused death and other causes death are presented in Fig. 2.
When subgroup analyses were conducted based on clinicopathological characteristics, results showed that the age, race, marital status, history of malignancy, primary site, grade, positive lymph nodes had no association (P > 0.05) with 5-year cumulative mortality. However, for cancer-specific deaths, the 5-year cumulative curves showed statistical differences in the subgroup of sex (P = 0.023), AJCC stage (P < 0.001), tumor size (P = 0.002), node status (P = 0.01), regional nodes examined (P < 0.001), bone metastasis (P < 0.001), liver metastasis (P < 0.001), lung metastasis (P < 0.001), receiving surgery (P < 0.001), receiving radiotherapy (P < 0.001), receiving chemotherapy (P = 0.047). It can be inferred that above mentioned covariates were associated with cancer-specific survival (P < 0.05). But not all of factors had significant associations with other reasons of deaths.

Multivariate SH model of ESRCC

To reduce the impact of competing risk bias, we performed the competing risk analysis with multivariate sub-distribution hazard function (Fine-Gray model) to quantify the effect of covariates on ESRCC (Table 2).
Table 2
Multivariate SH model in the training set and the validation set
Characteristics
Training Cohort (N = 282)
Validation Cohort (N = 282)
SHR
95% CI
P value
SHR
95% CI
P value
Age
 ≤ 65
Reference
  
Reference
  
 > 65
0.93
0.676–1.28
0.66
1.139
0.823–1.576
0.43
Sex
 Female
Reference
  
Reference
  
 Male
2.309
1.259–4.236
0.007
1.641
0.893–3.017
0.11
Race
 Others
Reference
  
Reference
  
 White
0.712
0.436–1.165
0.18
1.164
0.683–1.985
0.58
Marital status
 Widowed/others
Reference
  
Reference
  
 Divorced/Separated
1.033
0.523–2.042
0.92
0.885
0.448–1.748
0.73
 Married
0.878
0.539–1.428
0.6
0.767
0.469–1.255
0.29
 Single/Unmarried
0.963
0.525–1.766
0.9
0.947
0.539–1.662
0.85
History of Malignancy
 Previously
Reference
  
Reference
  
 Newly
1.897
0.652–5.52
0.24
0.715
0.284–1.802
0.48
Primary Site
 Others
Reference
  
Reference
  
 15-32 cm
1.244
0.625–2.476
0.53
1.55
0.527–4.588
0.43
 33-40 cm
0.647
0.376–1.113
0.12
2.612
1.182–5.772
0.018
Grade
 I-II
Reference
  
Reference
  
 III-IV
1.143
0.485–2.696
0.76
1.314
0.716–2.412
0.38
 Others
1.252
0.5–3.134
0.63
1.184
0.592–2.37
0.63
AJCC Stage
 I
Reference
  
Reference
  
 II
1.564
0.678–3.609
0.29
2.069
0.977–4.38
0.058
 III
1.899
0.769–4.686
0.16
4.336
1.883–9.987
< 0.001
 IV
2.358
0.99–5.614
0.053
3.173
1.435–7.017
0.004
Tumor Size
 T1
Reference
  
Reference
  
 T2
1.117
0.599–2.085
0.73
0.676
0.337–1.355
0.27
 T3
0.799
0.489–1.306
0.37
0.388
0.218–0.691
0.001
 T4
0.819
0.439–1.529
0.53
0.441
0.235–0.827
0.011
 TX
0.608
0.319–1.157
0.13
0.65
0.32–1.32
0.23
Node Status
 N0
Reference
  
Reference
  
 N1
1.103
0.685–1.775
0.69
0.71
0.444–1.136
0.15
 N2
0.952
0.51–1.779
0.88
0.548
0.285–1.054
0.072
 N3/NX
1.128
0.54–2.354
0.75
0.588
0.251–1.376
0.22
Regional Nodes Examined
 0
Reference
  
Reference
  
 0–10
1.286
0.653–2.533
0.47
0.962
0.297–3.112
0.95
 11–60
1.007
0.482–2.106
0.98
0.408
0.128–1.304
0.13
Positive Lymph Nodes
 0
Reference
  
Reference
  
 0–2
1.006
0.523–1.936
0.99
1.92
0.809–4.588
0.14
 3–35
1.537
0.706–3.346
0.28
2.949
0.94–9.253
0.064
Bone Metastases
 NO/Unknown
Reference
  
Reference
  
 YES
1.105
0.614–1.988
0.74
2.125
1.215–3.717
0.008
Liver Metastases
 NO/Unknown
Reference
  
Reference
  
 YES
1.573
0.848–2.919
0.15
2.238
1.301–3.85
0.004
Lung Metastases
 NO/Unknown
Reference
  
Reference
  
 YES
2.364
1.187–4.709
0.014
1.552
0.743–3.241
0.24
Surgery
 NO
Reference
  
Reference
  
 YES
0.497
0.271–0.91
0.024
0.331
0.116–0.948
0.039
Radiotherapy
 NO
Reference
  
Reference
  
 YES
1.098
0.616–1.956
0.75
1.991
1.014–3.906
0.045
Chemotherapy
 NO
Reference
  
Reference
  
 YES
0.82
0.498–1.351
0.44
0.757
0.491–1.168
0.21
In the training set (N = 282), sex (HR = 2.309, P = 0.007) and lung metastases (HR = 2.364, P = 0.0014) were independent risk factors associated with CSS, while receiving surgery (HR = 0.497, P = 0.024) was a protective factor correlated with better prognosis. Other covariates had no apparent associations with ESRCC (P > 0.05). Also, in the validation set (N = 282), the primary tumor located at 33–40 cm from incisor (HR = 2.612, P = 0.018), AJCC stage III (HR = 4.336, P < 0.001) and stage IV (HR = 3.173, P = 0.004), bone metastasis (HR = 2.125, P = 0.008), liver metastases (HR = 2.238, P = 0.004), receiving radiotherapy (HR = 1.991, P = 0.045) had a correlation with high risk of cancer-specific deaths, while T3 (HR = 0.388, P = 0.001), T4 (HR = 0.441, P = 0.011), and receiving surgery (HR = 0.331, P = 0.039) were associated with superior prognosis for ESRCC patients. The independent prognostic factors were not exactly the same from two cohorts owing to the heterogeneity of death events. But both two sets confirmed that protective surgery might be effective measures for ESRCC to achieve better prognosis.

Construction of a nomogram model

The variable selection was performed by stepwise regression. We constructed various models with different combinations of prognostic factors. Following the minimum Bayesian Information Criterions (BIC) principle, we chose the best fitting model and the corresponding variables were included. Explicitly, sex, surgery, lung metastases, liver metastases were identified as the significant variables. Based on the estimated coefficients, surgery acted as a protective factor (coefficient = -0.654), while male (coefficient = 0.708), liver metastasis (coefficient = 0.588), and lung metastasis (coefficient = 0.942) were independent risk factors.
A competing risk nomogram model (Fig. 3) was built with the weighted score and the variables mentioned above. The specific value for each factor was labeled with a straight line drawn upwards to the “point axis”. The total score could be calculated by adding up the points of all variables. We could easily estimate the probability of 5-year, 3-year, and 1-year ESRCC-specific survival by locating the “total point axis” down straight to the “survival axis”.

Validation of a nomogram model

For validation of the accuracy and stability of nomogram, we built two models for comparison. The first was a model composed of all covariates (ALL-model), and the second was a nomogram model based on BIC screening (BIC-model). We explored whether the BIC-based nomogram displayed better discrimination, calibration, and clinical benefits than the ALL-model. The nomogram was constructed with the data from the training set, while the validation set was used to validate the nomogram.

Discrimination and calibration

The nomogram model showed adequate discrimination with C-indexes of 0.61, 0.75, and 0.70 respectively for 5-year, 3-year, and 1-year CSS prediction. Receiver operating characteristic (ROC) curves were plotted in Fig. 4. The Area Under the Curve (AUC) was used to judge the accuracy of the prediction model. In the training set, the AUCs of nomogram (Fig. 4B) and All-model (Fig. 4A) were respectively 0.709 and 0.795 for 1-year, 0.757 and 0.84 for 3-year, and 0.642 and 0.74 for 5-year. Evidently, the nomogram had better prediction accuracy for 1- and 5-year survival probability (AUC > 0.7) than that of 3-year (0.5 < AUC < 0.7). However, all AUCs of nomogram were slightly smaller than the corresponding values of ALL-model. Similar outcomes could be observed in the testing set (Fig. 4C, D).
The stability of the nomogram was validated using the calibration curves in the training set (Fig. 4E, F) and validation set (Fig. 4G, H). Compared with the calibration curves of ALL-model (Fig. 4E, G), the calibration plots within BIC-model (Fig. 4F, H) displayed high consistency between the actual values and estimated probability for 12-month, 36-month, and 60-month survival, respectively.

Comprehensive evaluation

The Brier scores in BIC model were smaller (12-month = 0.225, 36-month = 0.214, 60-month = 0.221) than the ALL model (12-month = 0.258, 36-month = 0.258, 60-month = 0.274), representing a good prediction of our nomogram.

Decision curve analysis

DCA presented a clinical “net benefit” for one or more predictive models compared to default interventions of treating all or no patients. Threshold probability (X-axis) is the minimum probability of which further strategy would be warranted. The “intervention for none” and “intervention for all” respectively served as negative and positive control group. When analyzing the DCA curves of 12-month, All-model performed a little better than the BIC-model in training set (Fig. 5A), whereas the opposite trend was observed in validation set (Fig. 5E). Also, there was no remarkable discrepancy regarding 36-month (Fig. 5B, F) and 60-month (Fig. 5C, G) DCA plots between two models. Overall, except for a small range of low performances of BIC-based nomogram in the training set (Fig. 5A, B, C), interventions (such as surgery) on ESRCC patients on the fundamentals of the prediction model would lead to increased clinical benefits. Furthermore, as shown in Fig. 5D and H, both two arms supported that nomogram for 1-year prediction model showed relatively more predictive values for clinical decision making.

Subgroup analysis

We plotted the cumulative incidence curves using the data of the whole group (N = 564) and performed subgroup analysis on each prognostic factor. The 6-year cumulative incidence of CSS showed similar outcomes in both males and females (P = 0.996) (Fig. 6A). However, patients with liver metastasis, lung metastasis, or non-surgery all had a higher cumulative incidence of CSS than compared patients with statistically significant differences (P < 0.001), which is consistent with the findings of the multivariate SH model (Fig. 6B-D).

Discussion

Given the competing risks event that existed, we retrospectively analyzed the SEER database and constructed a nomogram model to evaluate independent prognostic factors and predict individualized 1-year, 3-year, and 5-year ESRCC-specific survival. This nomogram model showed a favorable discriminative ability, prediction accuracy, and clinical availability in both training and validation cohorts.
The incidence of esophageal adenocarcinoma has significantly increased in the western countries [15]. Especially, ESRCC was anticipated to account for 3–3.5% of all esophageal cancers [68]. This rare cancer was characterized by several clinicopathological features, including age, gender, race, material status, primary site, tumor grade, tumor stage, regional lymph nodes examined, positive lymph nodes, distant metastases, treatment modalities. There are numerous studies have spotlighted on the prediction model for esophageal adenocarcinoma. For instance, one study [16] established and validated a nomogram to predict individual survival of esophageal adenocarcinoma with six variables, including tumor size, T, N, M, age, grade. Also, its nomogram presented superior risk-stratifying capability than AJCC TNM staging system.
However, a majority of them performed traditional methods to analyze the survival data, such as Kaplan–Meier curve and Cox regression model, both of which are only suitable for single time-to-event outcome. Regarding time-to-event analysis, multiple reasons can lead to death, especially for patients with a long survival time. Notably, the occurrence of one kind of death might hinder the observation of other sorts of deaths [17]. For example, one study suggested that non-lung-cancer-specific death would confound the prediction of lung-cancer-specific death. Such a bias would increase along with age [18, 19]. Therefore, the risk of interest events might be overestimate by Kaplan–Meier method and the hazard ratio would be mistakenly evaluated by Cox regression model [20]. The competing risks caused by non-ESRCC might be ignored and censored. As reported, [21] if the percentage of competing event exceeded 10%, the Kaplan–Meier curve and Cox regression model would lead to severe result biases. Consequently, introducing CSS with competing risk model when predicting the prognosis of ESRCC patients is particularly essential.
To address above issues, competing risk model, a proportional sub-distribution hazards (SH) regression model, has been frequently conducted in survival analysis. But few researches focused on ESRCC-specific survival. To fulfill the research gaps, we estimated 5-year cumulative incidence of ESRCC in the SEER database from 2010 to 2015 and constructed a well-calibrated prognostic nomogram to predict 1-year, 3-year, and 5-year CSS. Consisting with prior studies of ESRCC, predictive parameters included the gender, lung metastases, liver metastases, and receiving surgery.
In the training cohort, gender made statistical difference in ESRCC-specific mortality. Male patients tended to have a poorer CSS than female patients (HR = 2.309, 95% CI: 1.259–4.236, P = 0.007). But the validation cohort showed there was no association between gender and prognosis in ESRCC patients (HR = 1.641, 95% CI: 0.893–3.017, P = 0.11). The selection bias might explain this paradoxical finding. Female patients were significantly less than male patients in our study, consistent with previous findings indicating a higher incidence in males [2226]. Sex-specific variants and androgen/estrogen balance associated with esophageal adenocarcinoma might improve the understanding of the reason for male predominance [27, 28]. Therefore, selection bias cannot be completely avoided owing to the different levels of disease exposure. With a limited enrollment of female patients, the median survival time is easily affected by individual difference. Overall, on the one hand, our nomogram model combined gender with other parameters showed a good discrimination and accurate prediction, supporting gender as a predictor for CSS. On the other hand, our finding indicated a poorer prognosis in men but conflicted with some studies manifesting no sex difference in prognosis observed for esophageal adenocarcinoma [2830]. Prospective studies are urgently required to further clarify the sex difference in prognosis.
Liver (coefficient = 0.588) and lung (coefficient = 0.94) distant metastases were two predominant predictors of this competing risk nomogram, both of which acted as independent risk variables for CSS. It is because distant metastases tend to indicate high malignancy and strong invasion ability of tumor, which is connected with limited treatment options, decreased survival, and worse prognosis. A real-world study [31] reported postoperative pathologic stage might be an independent factor associated with distant metastases (P = 0.004, HR = 0.15, 95% CI: 0.041–0.552). Particularly, T0-T2 stage had a lower risk of metastases (P = 0.011, HR = 0.119, 95% CI: 0.023–0.610). Consistently, in our multivariate SH model, T3 (P < 0.001, HR = 4.336, 95% CI: 1.883–9.987) and T4 (P = 0.004, HR = 3.173, 95% CI: 1.435–7.017) stage showed a higher risk of cancer-specific mortality in the validation set. But in the training set, the wide 95% CI and P-value were unsatisfied. It is possible that T stage functions as a risk factor for distant metastasis but contributes less to ESRCC-specific survival.
Surgery (coefficient = -0.654) is the only protective factor determined in the final competing risk nomogram model. Our study suggested ESRCC patients without surgery tend to have a shorter CSS and worse prognosis than those receiving resection. Both multivariate SH analysis and cumulative incidence curves supported this finding. As known, [32] surgery is the primary option for esophageal tumor, even though only 34.6% of patients with esophageal adenocarcinoma received complete resection. Surgical methods include esophagectomy only and esophagectomy plus other excisions. A prior study supported the OS (HR = 0.366, P < 0.001) and CSS (HR = 0.36, P < 0.001) of esophageal adenocarcinoma patients who underwent surgery were significantly longer than those without surgery, which is exactly consistent with our prediction model [32]. However, a retrospective analysis [33] put forward an interesting finding. Despite receiving modern induction therapy and surgery, the five-year OS of patients with ESRCC was only 31.3%, considerably less than historically reported for esophageal adenocarcinoma. We could infer that esophagectomy is an independent protective factor for ESRCC-specific survival, although it brings fewer clinical benefits to ESRCC patients than those without the SRC signature. Notably, either chemotherapy or radiotherapy showed limited prognostic values affecting CSS in our study. Due to the lack of enough evidence, the current standard care for ESRCC is still debatable [34]. Whether advanced ESRCC patients should firstly undergo surgery resection or multimodality therapy is still unclear. However, our study provided necessary evidence that surgery could significantly improve CSS for operable ESRCC patients. It highlighted the need to increase exposure to esophagectomy during early clinical decision-making. Some patients might be unresectable during planned esophagectomy or progressed during induction therapy [33], so surgical resection should be performed as early as possible after initial diagnosis. Novel biomarkers are urgently required to predict the efficacy of esophagectomy. Further clinical evidence-based data are required to compare the benefits of chemoradiation plus surgery versus surgery alone in prospective studies.
Overall, signet-ring-cell histology is a rare subtype of esophageal adenocarcinoma, which is known to have less well response to induction therapy and have decreased OS compared to patients with non-SRC histology [9]. With the increasing incidence of ESRCC, especially for western countries, more attention should be paid to construct prognosis prediction model. Since ESRCC are commonly associated with many complications, there are a sum of possibilities leading to biased results. Therefore, a competing risk model is of particular significance. To our best knowledge, this is the first study building the competing risk nomogram for prognosis prediction of patients with ESRCC. The performance of this nomogram appears to have effective discrimination ability, adequate model calibration, and a satisfying clinical net benefit. Our study provides several significant implications for clinical practice. Firstly, based on this large population-based study, it is necessary to increase exposure to resection in the early clinical decision-making, which may bring more benefits to ESRCC patients compared to other treatment strategies. Secondly, our nomogram model is an efficient approach to predict 1-, 3-, and 5-year CSS for patients with ESRCC. All variables could be easily obtained from clinical work. By calculating the score of each feature, this model could assist clinicians to make an accurate, comprehensive, and quick prognosis judgement and develop individualized therapy for each ESRCC patient. Thirdly, this competing risk model is an economic tool to avoid medical overuse, consisting of overdiagnosis and overtreatment [3537]. For unresectable ESRCC patients accompanying with more than one risk factor, overtreatment may place patients at risk of unnecessary adverse events. It is therefore clinicians could adopt palliative symptomatic treatment, aiming to alleviate patients’ pain and release economic burdens.
Our study has several outstanding advantages. First, our study is based on the large sample size provided by SEER database, ensuring the nomogram model robust enough. Second, cumulative incidence curve and multivariate SH model were employed to precisely estimate CSS by controlling the competing events. Third, we conducted decision curve analysis to reflect net benefit of the nomogram model, which is usually ignored in other similar reports. Meanwhile, the study has some limitations that deserves discussion. First, some recognized prognostic factors (such as genetic mutations) were not available in the SEER database. We failed to involve these parameters in our competing risk analysis. Second, since ESRCC is extremely rare in China, we lack enough data from the real world to make external validation. Third, most patients in our study were white and came from American. The universality of our competing risk model might be restricted. Finally, we only performed the internal validation for the predictive model, additional external validation is also required to guarantee its reliability and suitability.

Conclusion

In conclusion, a competing risk nomogram for ESRCC was successfully constructed and internally validated. This model is expected to predict 1-year, 3-year, and 5-year cancer-specific survival, and help oncologists and pathologists in clinical decision making and health care management for ESRCC patients. Future studies of ESRCC connected to molecular mutations, are vital as well for developing individualized therapy and improving cancer-specific survival.

Acknowledgements

None.

Declarations

This study follows the principles of the Declaration of Helsinki. The need for informed consent was eliminated since the SEER (Surveillance, Epidemiology, and End Results) database is freely accessible online.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
10.
Zurück zum Zitat Yoon HH, Khan M, Shi Q, Cassivi SD, Wu TT, Quevedo JF, et al. The prognostic value of clinical and pathologic factors in esophageal adenocarcinoma: a mayo cohort of 796 patients with extended follow-up after surgical resection. Mayo Clin Proc. 2010;85(12):1080–9. https://doi.org/10.4065/mcp.2010.0421. Epub 2010/12/03 PubMed PMID: 21123634; PubMed Central PMCID: PMCPMC2996151.CrossRefPubMedPubMedCentral Yoon HH, Khan M, Shi Q, Cassivi SD, Wu TT, Quevedo JF, et al. The prognostic value of clinical and pathologic factors in esophageal adenocarcinoma: a mayo cohort of 796 patients with extended follow-up after surgical resection. Mayo Clin Proc. 2010;85(12):1080–9. https://​doi.​org/​10.​4065/​mcp.​2010.​0421. Epub 2010/12/03 PubMed PMID: 21123634; PubMed Central PMCID: PMCPMC2996151.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Eguchi T, Bains S, Lee MC, Tan KS, Hristov B, Buitrago DH, et al. Impact of Increasing Age on Cause-Specific Mortality and Morbidity in Patients With Stage I Non-Small-Cell Lung Cancer: A Competing Risks Analysis. J Clin Oncol. 2017;35(3):281–90. https://doi.org/10.1200/jco.2016.69.0834. Epub 2017/01/18 PubMed PMID: 28095268; PubMed Central PMCID: PMCPMC5456376.CrossRefPubMed Eguchi T, Bains S, Lee MC, Tan KS, Hristov B, Buitrago DH, et al. Impact of Increasing Age on Cause-Specific Mortality and Morbidity in Patients With Stage I Non-Small-Cell Lung Cancer: A Competing Risks Analysis. J Clin Oncol. 2017;35(3):281–90. https://​doi.​org/​10.​1200/​jco.​2016.​69.​0834. Epub 2017/01/18 PubMed PMID: 28095268; PubMed Central PMCID: PMCPMC5456376.CrossRefPubMed
20.
Zurück zum Zitat de Glas NA, Kiderlen M, Vandenbroucke JP, de Craen AJ, Portielje JE, van de Velde CJ, et al. Performing Survival Analyses in the Presence of Competing Risks: A Clinical Example in Older Breast Cancer Patients. J Natl Cancer Inst. 2016;108(5):djv366. https://doi.org/10.1093/jnci/djv366. Epub 2015/11/29 PubMed PMID: 26614095.CrossRefPubMed de Glas NA, Kiderlen M, Vandenbroucke JP, de Craen AJ, Portielje JE, van de Velde CJ, et al. Performing Survival Analyses in the Presence of Competing Risks: A Clinical Example in Older Breast Cancer Patients. J Natl Cancer Inst. 2016;108(5):djv366. https://​doi.​org/​10.​1093/​jnci/​djv366. Epub 2015/11/29 PubMed PMID: 26614095.CrossRefPubMed
22.
Zurück zum Zitat Mathieu LN, Kanarek NF, Tsai HL, Rudin CM, Brock MV. Age and sex differences in the incidence of esophageal adenocarcinoma: results from the Surveillance, Epidemiology, and End Results (SEER) Registry (1973–2008). Dis Esophagus. 2014;27(8):757–63. https://doi.org/10.1111/dote.12147. Epub 2013/10/15 PubMed PMID: 24118313; PubMed Central PMCID: PMCPMC3979505.CrossRefPubMed Mathieu LN, Kanarek NF, Tsai HL, Rudin CM, Brock MV. Age and sex differences in the incidence of esophageal adenocarcinoma: results from the Surveillance, Epidemiology, and End Results (SEER) Registry (1973–2008). Dis Esophagus. 2014;27(8):757–63. https://​doi.​org/​10.​1111/​dote.​12147. Epub 2013/10/15 PubMed PMID: 24118313; PubMed Central PMCID: PMCPMC3979505.CrossRefPubMed
25.
31.
Zurück zum Zitat Du R, Ming J, Geng J, Zhu X, Zhang Y, Li S, et al. Establishment of prognostic models for adenocarcinoma of oesophagogastric junction patients with neoadjuvant chemoradiotherapy: a real-world study. Radiat Oncol (London, England). 2022;17(1):45. https://doi.org/10.1186/s13014-022-02016-3. Epub 2022/03/05 PubMed PMID: 35241109; PubMed Central PMCID: PMCPMC8896317.CrossRef Du R, Ming J, Geng J, Zhu X, Zhang Y, Li S, et al. Establishment of prognostic models for adenocarcinoma of oesophagogastric junction patients with neoadjuvant chemoradiotherapy: a real-world study. Radiat Oncol (London, England). 2022;17(1):45. https://​doi.​org/​10.​1186/​s13014-022-02016-3. Epub 2022/03/05 PubMed PMID: 35241109; PubMed Central PMCID: PMCPMC8896317.CrossRef
33.
34.
Zurück zum Zitat Kumar NAN, Jose A, Usman N, Rajan K, Munisamy M, Shetty PS, et al. Signet ring cell cancer of stomach and gastro-esophageal junction: molecular alterations, stage-stratified treatment approaches, and future challenges. Langenbeck’s Arch Surg. 2022;407(1):87–98. https://doi.org/10.1007/s00423-021-02314-6. Epub 2021/09/11 PubMed PMID: 34505199; PubMed Central PMCID: PMCPMC8847240.CrossRef Kumar NAN, Jose A, Usman N, Rajan K, Munisamy M, Shetty PS, et al. Signet ring cell cancer of stomach and gastro-esophageal junction: molecular alterations, stage-stratified treatment approaches, and future challenges. Langenbeck’s Arch Surg. 2022;407(1):87–98. https://​doi.​org/​10.​1007/​s00423-021-02314-6. Epub 2021/09/11 PubMed PMID: 34505199; PubMed Central PMCID: PMCPMC8847240.CrossRef
Metadaten
Titel
Prognosis prediction in esophageal signet-ring-cell carcinoma: a competing risk analysis
verfasst von
Chen Chen
Zehua Wang
Yanru Qin
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Gastroenterology / Ausgabe 1/2023
Elektronische ISSN: 1471-230X
DOI
https://doi.org/10.1186/s12876-023-02818-z

Weitere Artikel der Ausgabe 1/2023

BMC Gastroenterology 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

„Jeder Fall von plötzlichem Tod muss obduziert werden!“

17.05.2024 Plötzlicher Herztod Nachrichten

Ein signifikanter Anteil der Fälle von plötzlichem Herztod ist genetisch bedingt. Um ihre Verwandten vor diesem Schicksal zu bewahren, sollten jüngere Personen, die plötzlich unerwartet versterben, ausnahmslos einer Autopsie unterzogen werden.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Schlechtere Vorhofflimmern-Prognose bei kleinem linken Ventrikel

17.05.2024 Vorhofflimmern Nachrichten

Nicht nur ein vergrößerter, sondern auch ein kleiner linker Ventrikel ist bei Vorhofflimmern mit einer erhöhten Komplikationsrate assoziiert. Der Zusammenhang besteht nach Daten aus China unabhängig von anderen Risikofaktoren.

Update Innere Medizin

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