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

Open Access 01.12.2023 | Research

Risk prediction of second primary malignancies in patients after rectal cancer: analysis based on SEER Program

verfasst von: Yong-Chao Sun, Zi-Dan Zhao, Na Yao, Yu-Wen Jiao, Jia-Wen Zhang, Yue Fu, Wei-Hai Shi

Erschienen in: BMC Gastroenterology | Ausgabe 1/2023

Abstract

Background

This study will focus on exploring the clinical characteristics of rectal cancer (RC) patients with Second Primary Malignancies (SPMs) and constructing a prognostic nomogram to provide clinical treatment decisions.

Methods

We determined the association between risk factors and overall survival (OS) while establishing a nomogram to forecast the further OS status of these patients via Cox regression analysis. Finally, we evaluated the performance of the prognostic nomogram to predict further OS status.

Results

Nine parameters were identified to establish the prognostic nomogram in this study, and, the C-index of the training set and validation set was 0.691 (95%CI, 0.662–0.720) and 0.731 (95%CI, 0.676–0.786), respectively. The calibration curve showed a high agreement between the predicted and actual results, and the receiver operating characteristic (ROC) curves verified the superiority of our model for clinical usefulness. In addition, the nomogram classification could more precisely differentiate risk subgroups and improved the discrimination of SPMs’ prognosis.

Conclusions

We systematically explored the clinical characteristics of SPMs after RC and constructed a satisfactory nomogram.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12876-023-02974-2.
Yong-Chao Sun and Zi-Dan Zhao contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
RC
Rectal Cancer
OS
Overall Survival
ROC
Receiver Operating Characteristic
IPC
Initial Primary Cancer
LNR
Lymph Node Removed
NRI
Nutrition Risk Index
IDI
Integrated Discrimination Improvement
DCA
Decision Curve Analysis
SEER
Surveillance, Epidemiology, and End Results

Introduction

Rectal cancer represents the eighth most frequent diagnosed malignancy and the tenth most common reason for cancer-related deaths globally in 2018, [1] with approximately 732,210 new cases and 339,022 fatalities in 2020 [2, 3]. Nowadays, due to the progress of early diagnosis, comprehensive treatment, and advances in cancer detection, the OS of RC patients has greatly improved [4]. For early-treated rectal cancer, the 5-year OS rate among patients could even reach 90% [5, 6]. However, second primary malignancies are threatening the lives of RC patients who underwent long-term survival [7]. Recently, A growing number of studies have been carried out to investigate the risk factors for the development of SPMs in specific tumors, such as lung cancer [8], prostate cancer [9], breast cancer [10], stomach cancer [11], and so on. The prevalence of SPMs in RC survivors has been reported in earlier studies is 4-8% higher than in the normal population [12]. Factors thought to be influencing this higher rate have been explored in several studies, related to the patient’s genetic factors, lifestyle, environmental risk factors, and cancer therapy [1315].
Nomogram have been identified as a simpler and more sophisticated clinical prediction tool for predicting individualized OS based on clinical characteristics and risk factors [1618]. We discover that it is extremely important to understand the incidence and prognosis of SPM patients for treatment providers and RC patients. Therefore, this study will concentrate on the risk factors for SPMs and will develop a nomogram to forecast the 1-, 3-, and 5-year OS of SPMs after RC.

Materials and methods

Data source

Methods Data were obtained from SEER Research Plus Date,18 Registries, Nov 2020 Sub(2000–2018) in the Surveillance, Epidemiology, and End Results (SEER) database(http:/ /seer.cancer.gov)using SEER* Stat version 8.4.0. Clinicopathological information was gathered including age, race, gender, SPMs site, tumor size, histological type of SPMs and RC, TNM stage, clinical stage, surgical history of SPMs and RC, chemotherapy, radiotherapy, marital status, follow-up time, latency between RC and SPMs, respectively.

Definition of SPMs

SPMs was defined as metachronous invasive solid cancer developing ≥ 6 months after initial primary cancer (IPC), under criteria of Warren and Gates as modified by the National Cancer Institute [19]. The SEER database listed the pathologic subtypes of IPC and SPMs. To better distinguish SPMs from primary and metastatic tumors, we defined SPMs as second malignancy and histological different from IPC with an incubation period of not less than 6 months. Likewise, SEER database provided key clinical information on “malignant tumors for patient” and the “sequence number” of the multiple primary malignancies. It could be used to identify patients with SPM and index the sequence of multiple malignancies.

Patient selection

The clinicopathological information of a total of 4374 patients with rectal cancer was obtained from the SEER database. The following were the inclusion criteria: (1) Diagnosed age was between 20 and 80 years. (2) Rectal cancer was discovered in patients between January 2004 and December 2013, and the follow-up period was at least 5 years; (3) Detailed survival data and follow-up information on patients should be provided. The following were the exclusion criteria: (1) Patients without pathological confirmation of the diagnosis; (2) Patients who only provided death certificate records or autopsy records; (3) Latency periods of fewer than 6 months between IPC and SPMs. Next, we screened for the same histological type as rectal cancer (N = 2536), wherein 1838 patients were still diagnosed with SPMs. Patients with unclear clinical data were excluded, including the patients who have no TNM stage (N = 403), unknown lymph node removed (LNR) and marital status (N = 639), and unknown clinical stage of RC (N = 55). Finally, the prognostic nomogram was created using the risk factors that were identified, which were integrated from the detailed clinical data of 741 SPM patients with rectal cancer. Then, the data of 741 patients were randomly split into a training set (N = 585) and a verification set (N = 156) at a ratio of 7:2. Meanwhile, the training and validation set were used for external and internal validation, respectively. The precise details of SPMs screening were shown in Fig. 1.

Statistical analysis

To investigated the relationship between clinicopathological variables and OS of SPMs, univariate and multivariate Cox regression analyses were performed to specify the risk factors. Next, significantly different risk factors were used to build a nomogram that accurately forecast the 1-, 3- and 5-year survival rates of SPM patients. To verified the performance of the nomogram we constructed, the C-index was used to assess the accuracy of the prediction results. Next, the calibration curve was created to evaluate the consistency between predicted and actual results while bootstrapping with 1000 resamples was used to assess discrimination and calibration. Then, survival predictions for 1-, 3-and 5-year were estimated using the ROC curve. In addition, the nutrition risk index (NRI) and integrated discrimination improvement (IDI) were used to evaluate the degree the of accuracy between the nomogram and the conventional AJCC staging system, And the clinical usefulness and benefits of the nomogram were estimated by the decision curve analysis (DCA) plots.
In this study, R software (version 4.1.2) and SPSS 25.0 were both used for all statistical analysis. All tests were two-way and P < 0.05 was considered statistically significant.

Results

Characteristics of patients

A total of 51,611 patients diagnosed with rectal cancer during 2004–2013 was obtained from the SEER database, of which 4,374 patients were diagnosed with cancer more than 6 months after the initial diagnosis of RC. To rule out caused recurrence and metastasis of RC, the patient’s data with the same histological type as RC was ruled out. Ultimately, a total of 1838 (3.56%) patients diagnosed with SPMs were identified. The results showed that the median interval between RC and SPMs diagnosis was 36 months and the median age at SPMs diagnosis was 67.5 years. By using original data obtained from the SEER database, 741 cases of SPMs were found. After removing those with unclear clinical information, more than 1% of the patients’ SPM sites and histological types were listed (Fig. 2), suggesting that the three most common sites for SPMs were the Lung and Bronchus (18.35%), Urinary Bladder (15.11%), and Breast (11.20%) (Table 1) (Table S1). The three most prevalent histological types for SPMs were Squamous Cell Neoplasms (21.32%), Adenomas and Adenocarcinomas (18.76%), Transitional Cell Papillomas and Carcinomas (15.11%) (Table 1) (Table S2).
Table 1
Site and Histology types of SPMs after RC that the top 20
Site of SPMs
N
%
Histology of SPMs
N
%
All
741
100.00%
ALL
741
100.00%
Lung and Bronchus
136
18.35%
Squamous Cell Neoplasms
158
21.32%
Urinary Bladder
112
15.11%
Adenomas and Adenocarcinomas
139
18.76%
Rectum
83
11.20%
Transitional Cell Papillomas and Carcinomas
112
15.11%
Melanoma of the Skin
63
8.50%
Cystic, Mucinous and Serous Neoplasms
91
12.28%
Sigmoid Colon
23
3.10%
Epithelial Neoplasms, NOS
67
9.04%
Prostate
21
2.83%
Nevi and Melanomas
64
8.64%
Soft Tissue including Heart
18
2.43%
Complex Mixed and Stromal Neoplasms
16
2.16%
Anus, Anal Canal and Anorectum
18
2.43%
Nhl - Mature B-Cell Lymphomas
14
1.89%
NHL - Extranodal
15
2.02%
Ductal and Lobular Neoplasms
13
1.75%
Esophagus
14
1.89%
Soft Tissue Tumors and Sarcomas, NOS
10
1.35%
Stomach
14
1.89%
Complex Epithelial Neoplasms
8
1.08%
Corpus Uteri
14
1.89%
Fibromatous Neoplasms
7
0.94%
Tonsil
13
1.75%
Acinar Cell Neoplasms
7
0.94%
Larynx
13
1.75%
Germ Cell Neoplasms
6
0.81%
Pancreas
12
1.62%
Oseous and Chondromatous Neoplasms
3
0.40%
Ascending Colon
11
1.48%
Lipomatous Neoplasms
3
0.40%
Kidney and Renal Pelvis
11
1.48%
Myomatous Neoplasms
3
0.40%
Cecum
11
1.48%
Basal Cell Neoplasms
3
0.40%
Tongue
9
1.21%
Mesothelial Neoplasms
3
0.40%
Ovary
9
1.21%
Nhl - Mature t and Nk-Cell Lymphomas
2
0.27%
Abbreviations: SPMs: second primary malignancies; RC: rectal cancer
Final enrollment for further analysis included 741 patients in total, both the training set (N = 585) and the validation set (N = 156) were randomly divided from the 741 patients. Meanwhile, there was no significant difference in clinical information by using the χ2 test (P > 0.05), including the site of SPMs, histology of SPMs, age, race, TNM stage, treatment information, tumor size, and grade of SPMs (Table 2). The training set was used to build the nomogram and verify the model internally, while the validation set was utilized for external validation.
Table 2
Clinicopathological characteristics of SPM patients with RC
Variables
Training set
 
Validation set
  
χ²
P value
(n = 585)
 
(n = 156)
    
 
N
%
N
%
   
Site of SPMs
     
0.75
0.980
 Lung and Bronchus
106
18.1
30
19.2
   
 Urinary Bladder
88
8.7
24
7.7
   
 Rectum
68
43.4
15
44.9
   
 Melanoma of the Skin
51
11.6
12
9.6
   
 Sigmoid Colon
18
3.1
5
3.2
   
 Others
254
15.0
70
15.4
   
Histology of SPMs
     
6.54
0.257
 Squamous Cell Neoplasms
130
22.2
28
17.9
   
 Adenomas and Adenocarcinomas
114
19.5
25
16.0
   
 Transitional Cell Papilloma and Carcinomas
90
15.4
22
14.1
   
 Cystic, Mucinous and Serous Neoplasms
64
10.9
27
17.3
   
 Nevi and Melanomas
51
8.7
13
8.3
   
 Others
136
23.2
41
26.3
   
Age(years)
     
0.79
0.853
 <60
152
26.0
42
26.9
   
 60–69
190
32.5
47
30.1
   
 70–79
214
36.6
61
39.1
   
 ≥ 80
29
5.0
6
3.8
   
Race
     
5.29
0.071
 White
484
82.7
127
81.4
   
 Black
65
11.1
12
7.7
   
 Others
36
6.2
17
10.9
   
Stage-T
     
2.81
0.590
 Ta
141
24.1
34
21.8
   
 T1
186
31.8
42
26.9
   
 T2
122
20.9
38
24.4
   
 T3
89
15.2
26
16.7
   
 T4
47
8.0
16
10.3
   
Stage-N
     
5.15
0.161
 N0
482
82.4
113
74.4
   
 N1
55
9.4
22
14.1
   
 N2
43
7.4
16
10.3
   
 N3
5
0.9
5
1.3
   
Stage-M
     
0.29
0.593
 M0
528
90.3
143
91.7
   
 M1
57
9.7
13
8.3
   
Stage-T of RC
     
2.71
0.608
 Ta
74
12.6
24
15.4
   
 T1
140
23.9
34
21.8
   
 T2
87
14.9
29
18.6
   
 T3
254
43.4
63
40.4
   
 T4
30
5.1
6
3.8
   
Stage-N of RC
     
3.72
0.155
 N0
437
74.7
106
67.9
   
 N1
110
18.8
34
21.8
   
 N2
38
6.5
16
10.3
   
Stage-M of RC
     
0.91
0.341
 M0
552
94.4
144
92.3
   
 M1
33
5.6
12
7.7
   
SPMs Surgical history
       
 Yes
422
72.1
113
72.4
 
0.005
0.941
 No
163
27.9
43
27.6
   
Surgical history of RC
     
0.35
0.554
 Yes
511
87.4
139
89.1
   
 No
74
12.6
17
10.9
   
Histology of RC
     
0.75
0.689
 Others
70
12.0
19
12.2
   
 Ade
436
74.5
120
76.9
   
 Cystic, Mucinous and Serous Neoplasms
79
13.5
17
10.9
   
SPMs radiation record
     
0.58
0.445
 Yes
151
25.8
45
28.8
   
 No
434
74.19
111
71.2
   
Radiation record of RC
     
0.83
0.362
 Yes
291
49.7
84
53.8
   
 No
294
50.3
72
46.2
   
SPMs chemotherapy record
     
0.96
0.328
 Yes
215
36.8
64
41.0
   
 No
370
63.2
92
59.0
   
Chemotherapy record of RC
     
1.06
0.303
 Yes
318
54.4
92
59.0
   
 No
267
45.6
64
41.0
   
SPMs tumor size(cm)
     
5.45
0.141
 0–3
394
67.4
97
62.2
   
 3–5
86
14.7
32
20.5
   
 5–10
78
13.3
16
10.3
   
 ≥ 10
27
4.6
11
7.1
   
SPMs grade
     
4.73
0.316
 Well
56
9.6
9
5.8
   
 Moderately
183
31.3
41
26.3
   
 Poorly
94
16.1
29
18.6
   
 Undifferentiated
45
7.7
15
9.6
   
 Unknown
207
35.4
62
39.7
   
Abbreviations: SPMs: second primary malignancies; RC: rectal cancer, Ade: adenomas and adenocarcinomas

Prognostic factors selection and nomogram construction

Univariate and multivariate Cox regression analysis was applied to reveal OS-related factors in SPMs. The results (Table 3) show that the OS of SPMs was a significantly higher risk with age, TNM stage, stage M of RC, SPMs surgical history, SPMs tumor size (P < 0.001) and site(P = 0.009), while the OS of SPMs was a significantly lower risk with chemotherapy and radiotherapy(P<0.001). Multivariate Cox regression analysis revealed that age, stage-M, stage-M of RC, and SPMs surgical history(P<0.001), stage-T(P = 0.003), and stage-N(P = 0.012) were independent predictive variables for SPMs survival. According to the results of univariate and multivariate Cox regression analysis, 9 parameters including the site, age, stage TNM, stage M of RC, SPMs surgical history, SPMs radiotherapy records, SPMs chemotherapy records, and SPMs tumor size were used to establish a nomogram for predicting 1-, 3-, and 5-year OS (Fig. 3). To use the nomogram more conveniently, each of these characteristics was allocated a particular point on the scale. A total point was received for the individual patients, followed by a summary of the points from each parameter. Then, the probability of OS occurrence after 1, 3, and 5 years was predicted by transferring the entire score to the nomogram’s total score table. As an example, the total point of all variables for an SPM patient diagnosed with 60 years in urinary bladder site of 5 cm Tumor size, T2N2M0, M0 of RC, having SPMs Surgery record and Radiation record, but no chemotherapy record was 135, which corresponded to 1-,3-, and 5- year OS rates of about 88.3%,62.5%, and 50.1%, respectively.
Table 3
Univariate and multivariate Cox analysis of SPMs patients after RC in the training and validation set
Variables
Univariate analysis
 
Multivariate analysis
 
HR
CI (95%)
P value
HR
CI (95%)
P value
Site of SPMs
  
0.009
  
0.108
 Lung and Bronchus
1.000
  
1.000
  
 Urinary Bladder
0.383
0.276–0.530
<0.001
1.651
0.735–3.713
0.225
 Rectum
0.507
0.366–0.701
<0.001
1.020
0.648–1.605
0.932
 Melanoma of the Skin
0.349
0.229–0.530
<0.001
0.217
0.029–1.630
0.137
 Sigmoid Colon
0.427
0.243–0.748
<0.001
0.854
0.450–1.621
0.629
 Others
0.522
0.410–0.665
<0.001
0.840
0.619–1.140
0.263
Age(years)
  
<0.001
  
<0.001
 <60
1.000
  
1.000
  
 60–69
1.431
1.099–1.863
0.008
1.422
1.074–1.883
0.014
 70–79
1.758
1.370–2.256
<0.001
1.713
1.297–2.263
<0.001
 ≥ 80
2.499
1.624–3.846
<0.001
2.801
1.763–4.450
<0.001
Stage-T
  
<0.001
  
0.003
 Ta
1.000
  
1.000
  
 T1
1.028
0.786–1.345
0.839
0.819
0.586–1.146
0.244
 T2
1.388
1.045–1.844
0.024
0.835
0.577–1.207
0.337
 T3
1.545
1.145–2.084
0.004
1.159
0.769–1.748
0.480
 T4
3.340
2.377–4.693
<0.001
1.390
0.898–2.153
0.140
Stage-N
  
<0.001
  
0.012
 N0
1.000
  
1.000
  
 N1
1.618
1.222–2.143
<0.001
0.926
0.660–1.299
0.655
 N2
2.313
1.676–3.192
<0.001
1.534
1.071–2.197
0.020
 N3
3.369
1.668–6.802
<0.001
2.011
0.923–4.380
0.079
Stage-M
  
<0.001
  
<0.001
 M0
1.000
  
1.000
  
 M1
3.748
2.849–4.931
<0.001
2.523
1.800-3.537
<0.001
Stage-T of RC
  
0.005
  
0.094
 Ta
1.000
  
1.000
  
 T1
0.847
0.610–1.176
0.322
0.841
0.593–1.192
0.330
 T2
1.036
0.730–1.471
0.843
1.106
0.740–1.653
0.623
 T3
1.132
0.841–1.524
0.415
1.009
0.682–1.495
0.963
 T4
2.028
1.295–3.178
0.002
1.917
1.145–3.211
0.013
Stage-N of RC
  
0.017
  
0.721
 N0
1.000
  
1.000
  
 N1
1.187
0.942–1.496
0.146
0.975
0.753–1.261
0.844
 N2
1.848
1.324–2.578
<0.0001
1.314
0.894–1.932
0.164
Stage-M of RC
  
<0.001
  
<0.001
 M0
1.000
  
1.000
  
 M1
3.828
2.747–5.336
<0.001
3.113
2.144–4.521
<0.001
SPMs surgical history
  
<0.001
  
<0.001
 Yes
1.000
  
1.000
  
 No
2.403
1.974–2.924
<0.001
2.056
1.552–2.725
<0.001
SPMs radiation record
  
<0.001
  
0.129
 Yes
1.000
  
1.000
  
 No
0.707
0.577–0.866
<0.001
1.217
0.949–1.560
0.122
SPMs chemotherapy record
  
<0.001
  
0.177
 Yes
1.000
  
1.000
  
 No
0.581
0.482-0.700
<0.001
0.874
0.682–1.120
0.287
SPMs tumor size(cm)
  
<0.001
  
0.140
 0–3
1.000
  
1.000
  
 3–5
1.616
1.262–2.069
<0.001
1.377
1.032–1.839
0.030
 5–10
2.005
1.544–2.602
<0.001
1.365
1.007–1.850
0.045
 ≥ 10
0.965
0.613–1.520
0.879
0.792
0.472–1.330
0.378
Abbreviations: SPMs: second primary malignancies; RC: rectal cancer

Performance and validation of the nomogram

To assessed the discriminative potential of the constructed nomogram in this study, C-index in the training set 0.691 (95% CI, 0.662–0.720) and validation set 0.731 (95% CI, 0.676–0.786) was calculated, indicating that the nomogram has moderate accuracy. To assessed the correctness of our model, calibration plots were utilized to verify the consistency of our prediction and actual outcomes. The 1-, 3-, and 5-year 0 S calibration curves fit well with the 45° diagonal, indicating an excellent performance of the nomogram (Fig. 4). Meanwhile, the time-dependent ROC curves at 1-,3-and 5-year illustrated that the nomogram was more accurate in predicting OS prognosis in the training set 0.79 (95%,0.73–0.85),0.74 (95, 0.69–0.78) and 0.74 (95%,0.69–0.78), and validation set 0.72 (95%CI,0.58–0.85),0.72 (95%CI,0.64–0.80), and 0.70 (95%,0.62–0.79) (Fig. 5), respectively.
As shown in Fig. 6, DCA curves showed that the nomogram could more accurately forecast the likelihood of OS occurring after 1, 3, and 5 years, which, in both groups, may offer greater net clinical advantages than the AJCC stage model. Furthermore, we utilized the NRI and IDI to compare the accuracy of the nomogram with the usual AJCC staging system (Table 4). In the training set, the NRI for 1-3- and 5-year OS were 0.247(95%CI 0.022–0.503), 0.445(95%CI 0.363–0.689) and 0.445(95%CI 0.363–0.689), while the NRI for 1-3- and 5-year OS were 0.247(95%, CI 0.024–0.506), 0.445(95%, CI 0.299–0.682) and 0.075(95%CI 0.400–0.720) in the validation set. Additionally, the INI for 1-3- and 5-year OS were 0.030(P<0.001),0.072(P<0.001), and 0.080(P<0.001) in the training set, and 0.068(P<0.001),0.131(P<0.001) and 0.141(P<0.001) in the validation set. The NRI and IDI results demonstrated that the accuracy of the nomogram to predict OS is much superior than the usual AJCC staging system.
Table 4
NRI and IDI of the nomogram and the traditional AJCC staging system in OS prediction for RC patients
 
NRI
  
IDI
  
1-Year
3-Year
5-Year
1-Year
3-Year
5-Year
Training set(N = 585)
 Estimate
0.247
0.445
0.508
0.030
0.072
0.080
 95%CI
0.022–0.503
0.363–0.689
0.385–0.682
   
 P value
   
<0.001
<0.001
<0.001
Validation set(N = 156)
 Estimate
0.247
0.445
0.508
0.068
0.131
0.141
 95%CI
0.024–0.506
0.299–0.682
0.400–0.720
   
 P value
   
<0.001
<0.001
<0.001
Finally, a risk score for each patient was calculated by nomogram with an establishment of risk stratification (Fig. 7). In both the training (Fig. 7A) and validation (Fig. 7B) sets, the Kaplan-Meier survival curves displayed remarkable statistical difference between high and low-risk individuals (p<0.001).

Discussion

As the incidence of SPMs increased significantly, recent developments in SPMs had heightened the need for research on the monitoring, prognosis, and treatment decisions for clinical and public health [20, 21]. To investigated the prognosis of SPMs following RC, 9 parameters including the site, age, stage TNM, stage M of RC, SPMs surgical history, radiotherapy records, chemotherapy records, and tumor size were analyzed, which were applied to create a new nomogram that forecasts the survival rate of SPM patients. Taken together, our research showed that nomograph is superior to the AJCC staging system in predicting the probability of OS after 1 year, 3 years, and 5 years in the training set and validation set.
In reviewing the literature, Du et al. [22] reported that the three most prevalent sites of SPMs were neoplasms of colorectum (SIR 1.59, 95%CI 1.38–1.83), corpus uteri (SIR 2.11, 95%CI 1.62–2.76), and small intestine (SIR 4.00, 95%CI 2.91–5.49) in recently mete-analysis. Xu et al. [23] showed that Patients with RC were more likely to develop malignant tumors in the thyroid, uterine body, colon, rectum, lung/ bronchus. The same as our research results showed that the three most popular sites for SPMs were the Lung and Bronchus (18.35%), Urinary Bladder (15.11%), and Breast (11.20%). Therefore, it is of great significance to regular and long-term monitoring of the Lung and Bronchus, Urinary Bladder, and Rectum, which was necessary for RC patients at high risk.
Among the 9 parameters included in our nomogram, Age was recognized important risk contributor for SPM patients [24, 25]. Liu et al. [26] reported that Age (50–59:HR 0.958, 95%CI 0.842 − 0.091; 60–100:HR 1.557, 95%1.370–1.747; 18–49 as a reference) by multivariate analysis were all correlated with OS (P<0.001). Similarly, Li et al. [27] noted that Age (≥ 73:HR 1.482,95%CI 1.048–2.152; <73 as a reference) by multivariate analysis were all correlated with OS(P = 0.045). After dividing age into four age groups to better explore the relationship between age and overall survival, the results indicate that Age (60–69:HR1.422,95%CI1.074-1.883;70–79:HR 1.713,95%1.297–2.263; ≥80:HR 2.801,95%11.763–4.450; <60 as a reference) by multivariate analysis were all correlated with OS (P < 0.001). The degradation of the physical state, terrible treatment sensitivity, and the worsening cancer stage in elderly patients may all be contributing factors to these results.
Likewise, multivariate analysis in our study revealed that N stage (N1:HR 0.926, 95%CI 0.660–1.299; N2:1.534 95%CI 1.071–2.197;N3:HR 2.011,95%CI 0.923–4.380; N0 as a reference) for SPM patients had statistically significant OS rates(P = 0.012). This is consistent with those the findings of previous work that the N stage was one of the most significant contributions to OS [28, 29]. This view is supported by Park et al. [30] who reported that patients had higher pathological N stage (N1:HR 1.182,95%CI 1.191–1.845, P<0.001; N2:2.344 95%CI 1.779–3.289, P<0.001; N0 as a reference) significantly associated with OS, suggesting that surveillance was more frequent. As noted by Song et al [31], the N stage was considered as a potential predictor by LASSO, whose classification contributes most to the prognosis of survival in the nomogram they constructed.
Nomogram as a suitable scoring tool for clinical research, it could integrate the effects of various prognostic factors and present the results intuitively. Compared with the current AJCC sixth edition, the nomogram we created demonstrates a noticeably stronger capacity for risk stratification of RC SPM patients. Meanwhile, it is straightforward to gather nine prognostic factors on SPM patients, match that data with the nomogram we created, and calculate the corresponding scores. We could convenient to obtain the 1-, 3-, and 5-year OS by adding and matching the nomogram. The nomogram could help patients’ contributions to information on survival, clinical decision-making guidance, and treatment allocation. For those patients at high risk, they need active therapeutic and close monitoring to improve their overall survival.
Several questions still remain unanswered at present. First, although this study is a retrospective study and strictly complies with the inclusion and exclusion criteria, potential selection bias may have occurred. Secondly, Due to the lack of data relating to chemotherapy protocols and dose, it is not possible to evaluate the effects of different protocols and dose on the onset of secondary cancer. Finally, although our predictive model performs well through internal validation, additional external validation with other populations is still required.

Conclusions

In summary, this study was conducted to describe the clinical characteristics of SPMs in RC survivors and 9 clinical parameters are chosen to create a nomogram to forecast the 1-, 3-, and 5-year OS of SPM patients. It was also shown that the model prediction for OS in SPM patients was superior to the SEER historic stage with RC. Taken together, our findings might provide clinical prognostic guidelines for SPM patients, whose actual efficiency should be further improved through larger research further.

Acknowledgements

We thank the people involved in running the SEER database within the Division of Cancer Control and Population Sciences at the US National Cancer Institute (Bethesda, MD, USA).

Declarations

Competing interests

The authors declare no competing interests.
SEER database belongs to public databases. The patients involved in the database have obtained ethical approval. Users can download relevant data for free for research and publish relevant articles. Our study is based on open source data, so there are no ethical issues.
Not applicable.
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.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
12.
Zurück zum Zitat Lee Y-T, Liu C-J, Hu Y-W, Teng C-J, Tzeng C-H, Yeh C-M, et al. Incidence of second primary malignancies following colorectal Cancer: a distinct pattern of occurrence between Colon and rectal cancers and Association of Co-Morbidity with Second Primary Malignancies in a Population-Based cohort of 98,876 patients in Taiwan. Med (Baltim). 2015;94(26):e1079. https://doi.org/10.1097/MD.0000000000001079.CrossRef Lee Y-T, Liu C-J, Hu Y-W, Teng C-J, Tzeng C-H, Yeh C-M, et al. Incidence of second primary malignancies following colorectal Cancer: a distinct pattern of occurrence between Colon and rectal cancers and Association of Co-Morbidity with Second Primary Malignancies in a Population-Based cohort of 98,876 patients in Taiwan. Med (Baltim). 2015;94(26):e1079. https://​doi.​org/​10.​1097/​MD.​0000000000001079​.CrossRef
14.
Zurück zum Zitat Andrieu N, Launoy G, Guillois R, Ory-Paoletti C, Gignoux M. Estimation of the familial relative risk of cancer by site from a french population based family study on colorectal cancer (CCREF study). Gut. 2004;53(9):1322–8.CrossRefPubMedPubMedCentral Andrieu N, Launoy G, Guillois R, Ory-Paoletti C, Gignoux M. Estimation of the familial relative risk of cancer by site from a french population based family study on colorectal cancer (CCREF study). Gut. 2004;53(9):1322–8.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Dixon LB, Balder HF, Virtanen MJ, Rashidkhani B, Männistö S, Krogh V, et al. Dietary patterns associated with colon and rectal cancer: results from the dietary patterns and Cancer (DIETSCAN) Project. Am J Clin Nutr. 2004;80(4):1003–11.CrossRefPubMed Dixon LB, Balder HF, Virtanen MJ, Rashidkhani B, Männistö S, Krogh V, et al. Dietary patterns associated with colon and rectal cancer: results from the dietary patterns and Cancer (DIETSCAN) Project. Am J Clin Nutr. 2004;80(4):1003–11.CrossRefPubMed
19.
Zurück zum Zitat Swaroop VS, Winawer SJ, Kurtz RC, Lipkin M. Multiple primary malignant tumors. Gastroenterology. 1987;93(4):779–83.CrossRefPubMed Swaroop VS, Winawer SJ, Kurtz RC, Lipkin M. Multiple primary malignant tumors. Gastroenterology. 1987;93(4):779–83.CrossRefPubMed
24.
Zurück zum Zitat Valentini V, van Stiphout RG, Lammering G, Gambacorta MA, Barba MC, Bebenek M, et al. Nomograms for predicting local recurrence, distant metastases, and overall survival for patients with locally advanced rectal cancer on the basis of european randomized clinical trials. J Clin Oncol. 2011;29(23):3163–72. https://doi.org/10.1200/jco.2010.33.1595.CrossRefPubMed Valentini V, van Stiphout RG, Lammering G, Gambacorta MA, Barba MC, Bebenek M, et al. Nomograms for predicting local recurrence, distant metastases, and overall survival for patients with locally advanced rectal cancer on the basis of european randomized clinical trials. J Clin Oncol. 2011;29(23):3163–72. https://​doi.​org/​10.​1200/​jco.​2010.​33.​1595.CrossRefPubMed
28.
Zurück zum Zitat van Gijn W, van Stiphout R, van de Velde CJH, Valentini V, Lammering G, Gambacorta MA, et al. Nomograms to predict survival and the risk for developing local or distant recurrence in patients with rectal cancer treated with optional short-term radiotherapy. Ann Oncol. 2015;26(5):928–35. https://doi.org/10.1093/annonc/mdv023.CrossRefPubMed van Gijn W, van Stiphout R, van de Velde CJH, Valentini V, Lammering G, Gambacorta MA, et al. Nomograms to predict survival and the risk for developing local or distant recurrence in patients with rectal cancer treated with optional short-term radiotherapy. Ann Oncol. 2015;26(5):928–35. https://​doi.​org/​10.​1093/​annonc/​mdv023.CrossRefPubMed
Metadaten
Titel
Risk prediction of second primary malignancies in patients after rectal cancer: analysis based on SEER Program
verfasst von
Yong-Chao Sun
Zi-Dan Zhao
Na Yao
Yu-Wen Jiao
Jia-Wen Zhang
Yue Fu
Wei-Hai Shi
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Gastroenterology / Ausgabe 1/2023
Elektronische ISSN: 1471-230X
DOI
https://doi.org/10.1186/s12876-023-02974-2

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.