Survival benefit of radiotherapy in metastatic esophageal cancer: a population-based study
Original Article

Survival benefit of radiotherapy in metastatic esophageal cancer: a population-based study

Xiangpan Li1, Huibo Zhang1, Xuemei Jia2, Liming Xu3, Huali Liu1, Liang Chen1, Qibin Song1, Zhouguang Hui4

1Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; 2Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China; 3Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300070, China; 4Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 10000, China

Contributions: (I) Conception and design: X Li, Q Song, Z Hui; (II) Administrative support: Q Song; (III) Provision of study materials or patients: X Li; (IV) Collection and assembly of data: H Zhang, X Jia, L Xu, H Liu, L Chen; (V) Data analysis and interpretation: X Li; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Qibin Song. Department of Oncology, Renmin Hospital of Wuhan University, No. 99, Zhangzhidong Road, Wuhan 430060, China. Email: qibinsong1@163.com; Zhouguang Hui. Department of Radiation Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 10000, China. Email: lxp008cn@sina.com.

Background: Population-based estimates of survival benefits of radiotherapy on metastatic esophageal cancer (EC) are lacking. The aim of this study was to analyze survival benefits of radiotherapy in patients with metastatic EC at the time of cancer diagnosis.

Methods: Patients with metastatic EC were selected from Surveillance, Epidemiology, and End Results databases. The covariates included radiotherapy status, age, sex, insurance, histological type, differentiation, metastatic sites (bone, brain, liver, lung), and chemotherapy. Propensity score matching model was used to reduce bias of patients’ selection. Median overall survival (OS) and cancer-specific survival (CSS) were compared and Cox regression analysis was performed.

Results: A total of 4,761 patients with metastatic EC met the selection criteria. It was found that radiotherapy significantly improved 2-year OS (P=0.020) and 2-year CSS (P=0.009) in matched patients. In the propensity score model (N=3,672), Cox regression analysis demonstrated that radiotherapy was an independent prognostic factor which associated with a longer OS (P<0.001) and esophageal cause-specific survival in matched patients (P<0.001). Additionally, age, sex, insurance status, differentiation, number of metastatic sites and chemotherapy were also found to be significantly associated with OS and CSS in matched patients.

Conclusions: The population-based study demonstrated that patients with metastatic EC might benefit from radiotherapy. This data supports the proposal to change the current management for patients with metastatic EC.

Keywords: Metastatic esophageal cancer (metastatic EC); radiotherapy; prognosis


Submitted Dec 04, 2018. Accepted for publication May 21, 2019.

doi: 10.21037/tcr.2019.06.15


Introduction

Esophageal cancer (EC) is one of the most fatal malignant tumors globally, with a 5-year survival rate of 15–25% (1-3). With the improvement in diagnostic techniques, there are more than 50% patients detected at metastatic stage with incurable metastatic disease at diagnosis. For metastatic EC with distant metastasis, systematic chemotherapy is recommended as the standard therapy, however, the overall survival (OS) is still poor. External beam radiotherapy has been performed in the management for EC as definitive, preoperative, postoperative, or palliative therapy combining with chemotherapy (4-7). For patients with metastatic EC, radiotherapy is used as a palliative treatment modality to relieve symptoms such as dysphagia and chest pain (8,9). However, the effect of radiotherapy on survival of patients with metastatic EC is unclear.

The purpose of this study was to assess the impact of radiotherapy on the OS of metastatic EC based on the data available in the Surveillance, Epidemiology, and End Results (SEER) database, attempting to provide a novel concept for the change of traditional treatment modality to metastatic EC.


Methods

Patients

The SEER database, one of the largest databases of oncology patients in the United States, includes cancer incidence, treatment, and survival information for approximately 30% of the US population. SEER*Stat software version 8.3.5 was used and SEER data between 1973 and 2014 [“Incidence-SEER 18 Regs Custom data (with additional treatment fields), Nov 2016 sub (1973–2014 varying)”] was chosen for this study. Adult patients diagnosed with EC who had metastatic diseases between 2010 and 2014 (n=5,912) were enrolled. Patients treated with beam radiation (combined with or without other type of radiotherapy) were included in the cohort. Patients with unknown radiotherapy data were excluded (n=65). Patients for whom the presence of follow-up was unknown were not included (n=7). In addition, patients who were presented with “N/A not first tumor” were excluded (within SEER database) (n=1,079). All authors did not have access to information that could identify individual participants (Figure 1).

Figure 1 Flow diagram.

Statistical analysis

The enrolled patients were divided into two groups, radiotherapy group and non-radiotherapy group, and were longitudinally classified by age, sex, insurance, histological type, differentiation, metastatic sites (the bone, brain, liver and lung), chemotherapy code. Absolute numbers and incidence proportions were calculated.

All statistical analyses were performed using the SPSS statistical software (version 22.0; IBM Corporation). Propensity score matching model was performed to reduce the bias of patients’ selection and obtain the balanced population of radiotherapy and non-radiotherapy group. The standardized differences for matched variables were less than 0.1. Chi-square test was used to identify the differences of two groups. Kaplan-Meier method was used to obtain survival information. Log-rank test and Cox regression analyses were implemented to evaluate covariates for OS and esophageal cause-specific survival (CSS). A value of P<0.05 were considered statistically significant for all analyses.


Results

A total of 4,761 patients were finally enrolled in this study, including 1,901 with radiotherapy and 2,870 without. The baseline features of the 4,761 eligible patients are provided in Table 1. The majority of the patients were 60 to 79 years old (55.1%), male (84.1%), insured (92.3%), white (84.9%), adenocarcinoma (64.5%), poorly differentiated (47.9%). As for metastatic sites (to the brain, bone, lung, and liver), 3178 (66.8%) of patients had 1 or 2 metastatic sites, 202 patients (4.2%) had 3 or 4 sites. There were 993 (20.9%) patients who had no metastasis in any site of the liver, brain, bone and lung.

Table 1

Clinical and pathological characteristics of enrolled metastatic esophageal cancer patients

Characteristics Before propensity score matching, n (%) After propensity score matching, n (%)
All patients (n=4,761) Radiotherapy P All patients (n=3,672) Radiotherapy P
Yes (n=1,901) No (n=2,860) Yes (n=1,836) No (n=1,836)
Age, year <0.001 0.229
   20–39 70 (1.5) 19 (1.0) 51 (1.8) 50 (1.4) 18 (1.0) 32 (1.7)
   40–59 1,559 (32.7) 670 (35.2) 889 (31.1) 1,257 (34.2) 636 (34.6) 621 (33.8)
   60–79 2,622 (55.1) 1,051 (55.3) 1,571 (54.9) 2,037 (55.5) 1,022 (55.7) 1,015 (55.3)
   ≥80 510 (10.7) 161 (8.5) 349 (12.2) 328 (8.9) 160 (8.7) 168 (9.2)
Sex 0.868 0.383
   Male 4,002 (84.1) 1,600 (84.2) 2,402 (84.0) 3,113 (84.8) 1,547 (84.3) 1,566 (85.3)
   Female 759 (15.9) 301 (15.8) 458 (16.0) 559 (15.2) 189 (15.7) 270 (14.7)
Insurance code 0.003 0.093
   Insured 4,396 (92.3) 1,783 (93.8) 2,613 (91.4) 3,421 (93.2) 1,723 (93.8) 1,698 (92.5)
   Uninsured 235 (4.9) 82 (4.3) 153 (5.3) 160 (4.4) 77 (4.2) 83 (4.5)
   Unknown 130 (2.7) 36 (1.9) 94 (3.3) 91 (2.5) 36 (2.0) 55 (3.0)
Race <0.001 0.868
   White 4,043 (84.9) 1,563 (82.2) 2,480 (86.7) 3,089 (84.1) 1,543 (84.0) 1,546 (84.2)
   Black 465 (9.8) 212 (11.2) 253 (8.8) 363 (9.9) 180 (9.8) 183 (10.0)
   Other 237 (5.0) 120 (6.3) 117 (4.1) 207 (5.6) 107 (5.8) 100 (5.4)
   Unknown 16 (0.3) 6 (0.3) 10 (0.3) 13 (0.4) 6 (0.3) 7 (0.4)
Histological type <0.001 0.002
   SCC 1,118 (23.5) 547 (28.8) 571 (20.0) 913 (24.9) 492 (26.8) 421 (22.9)
   ADC 3,072 (64.5) 1,174 (61.8) 1,898 (66.4) 2,370 (64.5) 1,164 (63.4) 1,206 (65.7)
   Other 330 (6.9) 134 (7.0) 196 (6.9) 243 (6.6) 134 (7.3) 109 (5.9)
   Unknown 241 (5.1) 46 (2.4) 195 (6.8) 146 (4.0) 46 (2.5) 100 (5.4)
Differentiation <0.001 0.147
   Poorly differentiated 2,282 (47.9) 929 (48.9) 1,353 (47.3) 1,801 (49.0) 901 (49.1) 900 (49.0)
   Moderately differentiated 1,261 (26.5) 542 (28.5) 719 (25.1) 961 (26.2) 518 (28.2) 443 (24.1)
   Well differentiated 102 (2.1) 48 (2.5) 54 (1.9) 84 (2.3) 46 (2.5) 38 (2.1)
   Undifferentiated 72 (1.5) 32 (1.7) 40 (1.4) 53 (1.4) 32 (1.7) 21 (1.1)
   Unknown 944 (19.8) 350 (18.4) 694 (24.3) 773 (21.1) 339 (18.5) 434 (23.6)
Bone metastasis <0.001 <0.001
   Yes 1,123 (23.6) 564 (29.7) 559 (19.5) 884 (24.1) 550 (30.0) 334 (18.2)
   No 3,437 (72.2) 1,278 (67.2) 2,159 (75.5) 2,659 (72.4) 1,227 (66.8) 1,432 (78.0)
   Unknown 201 (4.2) 59 (3.1) 142 (5.0) 129 (3.5) 59 (3.2) 70 (3.8)
Brain metastasis <0.001 <0.001
   Yes 256 (5.4) 181 (9.5) 75 (2.6) 226 (6.2) 181 (9.9) 45 (2.5)
   No 4,269 (89.7) 1,652 (86.9) 2,617 (91.5) 3,291 (89.6) 1,587 (86.4) 1,704 (92.8)
   Unknown 236 (5.0) 68 (3.6) 168 (5.9) 155 (4.2) 68 (3.7) 87 (4.7)
Liver metastasis <0.001 <0.001
   Yes 2,270 (47.7) 675 (35.5) 1,595 (55.8) 1,652 (45.0) 666 (36.3) 986 (53.7)
   No 2,321 (48.8) 1,170 (61.5) 1,151 (40.2) 1,906 (51.9) 1,115 (60.7) 791 (43.1)
   Unknown 170 (3.6) 56 (3.0) 114 (4.0) 114 (3.1) 55 (3.0) 59 (3.2)
Lung metastasis 0.001 0.291
   Yes 1,375 (28.9) 526 (27.7) 849 (29.7) 1,037 (28.2) 510 (27.8) 527 (28.7)
   No 3,132 (65.8) 1,297 (68.2) 1,835 (64.2) 2,470 (67.3) 1,249 (68.0) 1,221 (66.5)
   Unknown 254 (5.3) 78 (4.1) 176 (6.2) 165 (4.5) 77 (4.2) 88 (4.8)
Metastatic sites to the brain, bone, lung, and liver <0.001 0.084
   0 993 (20.9) 487 (25.6) 506 (17.7) 828 (22.5) 456 (24.8) 372 (20.3)
   1–2 3,178 (66.8) 1,196 (62.9) 1,982 (69.3) 2,449 (66.7) 1,163 (63.3) 1,286 (70.0)
   3–4 202 (4.2) 104 (5.5) 98 (3.4) 142 (3.9) 104 (5.7) 38 (2.1)
   Unknown 388 (8.1) 114 (6.0) 274 (9.6) 253 (6.9) 113 (6.2) 140 (7.6)
Chemotherapy <0.001 <0.001
   Yes 2,880 (60.5) 1,403 (73.8) 1,477 (51.6) 2,494 (67.9) 1,338 (72.9) 1,156 (63.0)
   No/unknown 1,881 (39.5) 498 (26.2) 1,383 (48.4) 1,178 (32.1) 498 (27.1) 680 (37.0)

SCC, squamous carcinoma; ADC, adenocarcinoma.

The median OS (mOS) and CSS (mCSS) for the entire cohort were 4.9 and 5.0 months, and the radiotherapy group was 7.0 and 6.9 months, while 3.0 and 4.0 months in the non-radiotherapy group, respectively (P<0.001) (Figure 2A,B). Univariate and multivariable Cox regression demonstrated that radiotherapy was significantly associated with longer mOS and mCSS (Tables 2,3). In addition, metastatic disease to 1–2 sites (P<0.001) or 3–4 metastatic sites (P<0.001) of the brain, bone, lung and liver, or greater than 80 years (P=0.002), uninsured status (P<0.001), and non-chemotherapy treatment (P<0.001) were significantly associated with poorer OS, while age 20 to 39 years (P=0.002), 40 to 59 years (P=0.002), 60 to 79 years (P=0.003), female (P<0.001), ADC (P=0.035), moderately differentiated (P<0.001) and well differentiated tumor (P=0.040) were significantly associated with better OS. Moreover, metastatic disease to 1–2 sites (P<0.001) or 3–4 sites (P<0.001), greater than 80 years old (P=0.016), uninsured status (P<0.001), non-chemotherapy treatment (P<0.001) were significantly associated with decreased CSS. Factors that were statistically associated with longer CSS in multivariable Cox regression analysis were female (P<0.001) and moderately differentiated tumor (P<0.001).

Figure 2 OS and CSS curves for the effect of radiotherapy. (A,B) Before propensity score matching; (C,D) after propensity score matching. OS, overall survival; CSS, cancer-specific survival.

Table 2

Univariate Cox regression analysis for OS and CSS

Characteristics Before propensity score matching After propensity score matching
OS CSS OS CSS
HR 95% CI P HR 95% CI P HR 95% CI P HR 95% CI P
Age, y
   ≥80 1.000 1.000 1.000 1.000
   60–79 0.665 0.601–0.735 <0.001 0.688 0.620–0.765 <0.001 0.714 0.629–0.809 0.001 0.753 0.660–0.860 <0.001
   40–59 0.604 0.543–0.673 <0.001 0.628 0.562–0.703 <0.001 0.644 0.564–0.735 <0.001 0.686 0.597–0.788 <0.001
   20–39 0.496 0.371–0.663 <0.001 0.531 0.396–0.712 <0.001 0.570 0.405–0.803 <0.001 0.617 0.435–0.875 <0.001
Sex
   Male 1.000 1.000 1.000 1.000
   Female 0.915 0.839–0.997 0.043 0.990 0.823–0.984 0.021 0.882 0.797–0.977 0.016 0.867 0.780–0.963 0.008
Insurance code
   Insured 1.000 1.000 1.000 1.000
   Uninsured 1.376 1.198–1.581 <0.001 1.386 1.202–1.598 <0.001 1.446 1.224–1.708 <0.001 1.444 1.217–1.714 <0.001
   Unknown 1.175 0.975–1.418 0.091 1.132 0.931–1.378 0.214 1.173 0.937–1.469 0.165 1.160 0.919–1.464 0.211
Race
   White 1.000 1.000 1.000 1.000
   Black 1.127 1.017–1.248 0.023 1.142 1.028–1.268 0.013 1.143 1.017–1.284 0.025 1.151 1.021–1.297 0.021
   Other 0.869 0.752–1.004 0.056 0.842 0.724–0.979 0.025 0.917 0.785–1.071 0.273 0.900 0.767–1.057 0.200
   Unknown 0.994 0.577–1.714 0.984 0.970 0.550–1.709 0.915 0.940 0.505–1.749 0.844 0.892 0.464–1.717 0.733
Histological type
   SCC 1.000 1.000 1.000 1.000
   ADC 0.881 0.818–0.949 0.001 0.880 0.815–0.950 0.001 0.865 0.787–0.929 <0.001 0.853 0.783–0.929 <0.001
   Other 1.059 0.928–1.209 0.396 1.070 0.934–1.225 0.330 1.081 0.927–1.261 0.318 1.083 0.926–1.268 0.318
   Unknown 1.576 1.364–1.821 <0.001 1.511 1.299–1.757 <0.001 1.512 1.264–1.810 <0.001 1.459 1.210–1.759 <0.001
Differentiation
   Poor 1.000 1.000 1.000 1.000
   Moderate 0.792 0.734–0.855 <0.001 0.798 0.738–0.862 <0.001 0.741 0.679–0.808 <0.001 0.753 0.689–0.823 <0.001
   Well 0.802 0.647–0.995 0.045 0.810 0.650–1.009 0.061 0.802 0.632–1.018 0.069 0.816 0.639–1.040 0.101
   Undifferentiated 1.030 0.800–1.326 0.820 1.054 0.815–1.364 0.686 1.010 0.753–1.354 0.947 1.048 0.779–1.410 0.759
   Unknown 0.964 0.891–1.044 0.373 0.958 0.882–1.040 0.303 0.880 0.802–0.966 0.007 0.877 0.796–0.965 0.007
Bone metastasis
   No 1.000 1.000 1.000 1.000
   Yes 1.391 1.293–1.495 <0.001 1.399 1.29801.507 <0.001 1.436 1.332–1.560 <0.001 1.143 1.326–1.571 <0.001
   Unknown 1.108 0.950–1.292 0.191 1.069 0.911–1.255 0.413 1.045 0.861–1.267 0.658 1.033 0.846–1.262 0.748
Brain metastasis
   No 1.000 1.000 1.000 1.000
   Yes 1.255 1.097–1.435 0.001 1.255 1.093–1.441 0.001 1.282 1.109–1.482 0.001 1.287 1.109–1.493 0.001
   Unknown 1.060 0.920–1.221 0.421 1.024 0.883–1.188 0.751 0.987 0.827–1.178 0.888 0.978 0.815–1.174 0.813
Liver metastasis
   No 1.000 1.000 1.000 1.000
   Yes 1.246 1.170–1.327 <0.001 1.265 1.185–1.350 <0.001 1.264 1.176–1.359 <0.001 1.282 1.190–1.381 <0.001
   Unknown 1.109 0.938–1.313 0.226 1.058 0.886–1.264 0.530 0.988 0.803–1.215 0.907 0.939 0.754–1.168 0.570
Lung metastasis
   No 1.000 1.000 1.000 1.000
   Yes 1.279 1.194–1.370 <0.001 1.274 1.187–1.368 <0.001 1.295 1.196–1.402 <0.001 1.294 1.193–1.404 <0.001
   Unknown 1.090 0.948–1.253 0.228 1.086 0.941–1.253 0.261 0.985 0.827–1.173 0.865 0.994 0.831–1.189 0.951
Metastatic sites to the brain, bone, lung, and liver
   0 1.000 1.000 1.000 1.000
   1–2 1.458 1.346–1.579 <0.001 1.473 1.357–1.600 <0.001 1.499 1.371–1.639 <0.001 1,157 1.384–1.664 <0.001
   3–4 2.268 1.932–2.664 <0.001 2.290 1.941–2.701 <0.001 2.253 1.945–2.847 <0.001 2.377 1.954–2.892 <0.001
   Unknown 1.556 1.378–1.780 <0.001 1.563 1.370–1.784 <0.001 1.435 1.229–1.676 <0.001 1.452 1.238–1.704 <0.001
Radiotherapy
   Yes 1.000 1.000 1.000
   No 1.407 1.320–1.499 <0.001 1.405 1.316–1.500 <0.001 1.231 1.146–1.322 <0.001 1.293 1.146–1.327 <0.001
Chemotherapy
   Yes 1.000 1.000 1.000 1.000
   No/unknown 3.367 3.153–3.596 <0.001 3.341 3.123–3.575 <0.001 3.331 3.083–3.600 <0.001 3.314 3.060–3.590 <0.001

OS, overall survival; CSS, cancer-specific survival.

Table 3

Multivariate Cox regression analysis for OS and CSS

Characteristics Before propensity score matching After propensity score matching
OS CSS OS CSS
HR 95% CI P HR 95% CI P HR 95% CI P HR 95% CI P
Age, y
   ≥80 1.000 1.000 1.000 1.000
   60–79 0.853 0.769–0.947 0.003 0.875 0.785–0.975 0.016 0.867 0.762–0.986 0.030 0.915 0.799–1.048 0.198
   40–59 0.837 0.749–0.937 0.002 0.859 0.765–0.966 0.011 0.835 0.727–0.958 0.010 0.889 0.769–1.027 0.111
   20–39 0.623 0.465–0.835 0.002 0.661 0.491–0.890 0.006 0.653 0.462–0.923 0.016 0.915 0.799–1.048 0.198
Sex
   Male 1.000 1.000 1.000 1.000
   Female 0.854 0.782–0.934 <0.001 0.840 0.766–0.920 <0.001 0.828 0.746–0.920 <0.001 0.815 0.731–0.908 <0.001
Insurance code
   Insured 1.000 1.000 1.000 1.000
   Uninsured 1.343 1.166–1.547 <0.001 1.332 1.151–1.540 <0.001 1.404 1.184–1.663 <0.001 1.387 1.165–1.652 <0.001
   Unknown 0.881 0.727–1.068 0.197 0.854 0.698–1.045 0.126 0.958 0.758–1.210 0.718 0.953 0.749–1.213 0.696
Race
   White 1.000 1.000 1.000
   Black 1.073 0.959–1.202 0.220 1.083 0.965–1.217 0.175 1.052 0.925–1.197 0.439 1.051 0.921–1.199 0.461
   Other 0.894 0.771–1.036 0.137 0.865 0.741–1.009 0.064 0.897 0.765–1.051 0.177 0.879 0.746–1.036 0.124
   Unknown 0.881 0.507–1.534 0.655 0.858 0.482–1.527 0.602 0.804 0.424–1.523 0.503 0.750 0.382–1.470 0.402
Histological type
   SCC 1.000 1.000 1.000 1.000
   ADC 0.920 0.852–0.994 0.035 0.925 0.848–1.009 0.078 0.933 0.848–1.026 0.151 0.924 0.838–1.018 0.110
   Other 1.019 0.889–1.169 0.787 1.048 0.908–1.210 0.521 1.130 0.959–1.331 0.144 1.132 0.957–1.340 0.149
   Unknown 1.258 1.081–1.464 0.003 1.221 1.038–1.435 0.016 1.330 1.101–1.607 0.003 1.285 1.055–1.564 0.013
Differentiation
   Poor 1.000 1.000 1.000 1.000
   Moderate 0.767 0.710–0.828 <0.001 0.772 0.713–0.835 <0.001 0.766 0.701–0.837 <0.001 0.778 0.711–0.853 <0.001
   Well 0.798 0.643–0.990 0.040 0.804 0.645–1.003 0.053 0.785 0.618–0.997 0.047 0.795 0.622–1.105 0.065
   Undifferentiated 0.855 0.660–1.109 0.238 0.888 0.681–1.158 0.380 0.809 0.594–1.102 0.179 0.845 0.618–1.155 0.290
   Unknown 0.798 0.735–0.867 <0.001 0.800 0.735–0.871 <0.001 0.772 0.702–0.849 <0.001 0.771 0.699–0.851 <0.001
Metastatic sites to the brain, bone, lung, and liver
   0 1.000 1.000 1.000 1.000
   1–2 1.357 1.251–1.471 <0.001 1.363 1.253–1.482 <0.001 1.414 1.291–1.548 <0.001 1.427 1.300–1.567 <0.001
   3–4 2.365 2.012–2.781 <0.001 2.379 2.014–2.810 <0.001 2.460 2.029–2.981 <0.001 2.477 2.032–3.019 <0.001
   Unknown 1.290 1.132–1.471 <0.001 1.291 1.127–1.478 <0.001 1.153 0.982–1.353 0.082 1.162 0.986–1.370 0.074
Radiotherapy
   Yes 1.000 1.000 1.000 1.000
   No 1.229 1.151–1.313 <0.001 1.227 1.147–1.313 <0.001 1.218 1.132–1.310 <0.001 1.219 1.131–1.314 <0.001
Chemotherapy
   Yes 1.000 1.000 1.000 1.000
   No/unknown 3.211 2.995–3.442 <0.001 3.204 2.982–3.441 <0.001 3.195 2.944–3.467 <0.001 3.204 2.946–3.485 <0.001

OS, overall survival; CSS, cancer-specific survival.

After propensity score matching, 3,672 of 4,761 patients were included (1,836 for each of radiotherapy or non-radiotherapy group) (Table 1) and also found that radiotherapy improved OS and CSS (P<0.001) (Figure 2C,D). Radiotherapy showed significant 2-year survival benefits in patients with age older than 80 years (2-year OS, P=0.048; CSS, P=0.018), male (2-year OS, P=0.020; CSS, P=0.011), white race (2-year OS, P=0.038; CSS, P=0.006), squamous carcinoma (2-year OS, P=0.002; CSS, P<0.001), poor differentiation (2-year OS, P=0.002; CSS, P<0.001), brain (2-year OS, P<0.001; CSS, P<0.001) metastasis, other sites (sites except for the brain, bone, lung, and liver, 2-year OS, P<0.001; CSS, P<0.001), however, there were no statistically significant survival differences in patients with bone and liver metastasis, 1–2 or 3–4 metastatic sites (Table 4). Interestingly, no survival difference was found between chemotherapy alone and radiotherapy combined with chemotherapy (2-year OS, P=0.177; CSS, P=0.080). Cox regression demonstrated that radiotherapy was an independent prognostic factor which was significantly associated with longer mOS and mCSS in matched patients (Tables 2,3). In addition, metastatic disease to 1–2 sites (P<0.001) or 3–4 metastatic sites (P<0.001) of the brain, bone, lung and liver, age greater than 80 years (P≤0.030), uninsured status (P<0.001), male (P<0.001), poor differentiation (P≤0.047), non-chemotherapy treatment (P<0.001) were associated with poorer OS. While metastatic disease to 1–2 sites (P <0.001) or 3–4 sites (P<0.001), uninsured status (P<0.001), male (P<0.001), poor differentiation (P<0.001), non-chemotherapy treatment (P<0.001) were significantly associated with decreased CSS.

Table 4

Z test for the effects of radiotherapy on OS and CSS in matched patients

Characteristics 2-year OS 2-year CSS
RT (%) Non-RT (%) Z P RT (%) Non-RT (%) Z P
RT 11.6 8.8 2.325 0.020 13.4 9.9 2.602 0.009
Age
   20–39 8.4 15.6 0.65 0.517 9.3 15.6 −0.54 1.410
   40–59 12.3 10.6 0.83 0.407 13.6 11.9 0.75 0.452
   60–79 11.5 8.4 1.90 0.057 13.2 9.3 2.29 0.022
   ≥80 9.1 2.9 1.98 0.048 13.8 4.2 2.37 0.018
Sex
   Male 10.9 8.1 2.33 0.020 12.5 9.1 2.53 0.011
   Female 15.1 13.6 0.43 0.665 17.7 15.4 0.59 0.554
Insurance code
   Insured 11.7 9.2 2.08 0.038 13.4 10.3 2.30 0.021
   Uninsured 8.2 0.0 2.28 0.023 10.2 0.0 2.43 0.015
   Unknown 15.3 7.5 1.00 0.320 16.7 8.1 1.01 0.312
Race
   White 12.0 8.7 2.45 0.014 13.6 9.7 2.74 0.006
   Black 7.0 8.9 0.60 0.551 8.8 9.4 0.17 0.865
   Other 13.8 10.3 0.65 0.515 17.0 12.5 0.73 0.465
   Unknown 20.0 0.0 1.12 0.264 40.0 0.0 1.826 0.068
Histological type
   SCC 12.1 5.6 3.04 0.002 13.8 6.0 3.51 0.000
   ADC 12.1 10.6 0.96 0.335 13.8 11.7 1.24 0.216
   Other 7.1 6.0 0.28 0.782 9.4 7.2 0.48 0.633
   Unknown 4.9 3.0 0.51 0.609 8.7 4.8 0.76 0.448
Differentiation
   Poor 12.1 5.6 3.04 0.002 10.8 7.1 3.51 0.000
   Moderate 12.1 10.6 0.96 0.335 18.2 12.3 2.14 0.032
   Well 7.1 6.0 0.28 0.782 16.4 8.2 1.03 0.301
   Undifferentiated 4.9 3.0 0.51 0.609 0.0 15.1 1.61 0.108
   Unknown 12.1 5.6 3.04 0.002 13.6 13.1 0.16 0.870
Bone metastasis
   Yes 4.2 4.6 0.26 0.798 5.1 5.5 0.20 0.841
   No 14.6 9.7 0.87 0.384 16.5 10.8 3.48 0.001
   Unknown 14.9 9.4 0.82 0.411 21.9 9.8 1.65 0.099
Brain metastasis
   Yes 8.1 0.0 3.52 0.000 10.7 0.0 3.82 0.000
   No 11.6 9.0 2.16 0.031 13.2 10.1 2.30 0.021
   Unknown 18.9 9.5 1.45 0.148 23.1 10.0 1.89 0.059
Liver metastasis
   Yes 7.0 8.7 1.04 0.296 8.4 9.7 0.71 0.481
   No 13.7 8.6 3.01 0.003 15.3 9.6 3.10 0.002
   Unknown 20.7 11.9 1.16 0.244 30.0 12.8 2.07 0.038
Lung metastasis
   Yes 8.3 7.0 0.66 0.511 9.7 8.3 2.27 0.023
   No 12.4 9.4 2.02 0.044 14.1 10.4 1.78 0.076
   Unknown 20.1 12.3 1.20 0.229 24.6 12.7 1.78 0.076
Metastatic sites to the brain, bone, lung, and liver
   0 19.5 9.0 3.80 0.000 21.2 9.9 3.80 0.000
   1–2 8.6 9.1 0.37 0.710 10.1 10.3 0.81 0.420
   3–4 2.6 0.0 1.44 0.149 3.8 0.0 1.52 0.129
   Unknown 17.2 7.7 2.00 0.045 21.1 8.2 2.51 0.012
Chemotherapy
   Yes 14.8 12.6 1.35 0.177 16.8 13.7 1.75 0.080
   No/unknown 2.8 2.2 0.55 0.579 3.5 2.7 0.59 0.556

OS, overall survival; CSS, cancer-specific survival.


Discussion

This is the first large population-based study evaluating the effect of radiotherapy in the management of metastatic EC based on the SEER database, revealing that radiotherapy was an independent prognostic factor associated with survival benefits of patients with metastatic EC. EC is one of the leading cause of cancer deaths worldwide with poor prognosis (3,10,11). In general, radiotherapy plays an important role in the treatment of local EC. A study reported a 5-year OS rate of 21% in 101 patients with locally EC receiving radiotherapy alone (12). Then chemoradiotherapy became the preferred treatment and had been shown to improve the quality of life and prolong survival for patients with local metastatic or unresectable EC. Systemic therapy is the standard treatment for metastatic disease, but symptoms caused by metastasis disease often require multidisciplinary management including radiotherapy. However, the value of radiotherapy in the treatment of metastatic EC has not yet been fully evaluated before. This present study can complement the treatment recommended in the current guidelines.

Although the research from Wu et al. (13) found that combining surgery with radiotherapy could improve survival in metastatic EC, it based on older populations, older methods of radiotherapy, and did not analyze the clinical benefits of chemotherapy combined with radiation therapy. A phase II study compared concurrent chemoradiation therapy (CCRT) with chemotherapy alone in stage IV esophageal squamous cell carcinoma, demonstrating that CCRT significantly prolonged median progression-free survival (mPFS, 9.3 vs. 4.7 months, P=0.021) and mOS (18.3 vs. 10.2 months, P=0.001) (14). This study challenged the status of standard treatment modality for metastatic EC treated with chemotherapy alone. The results showed that metastatic EC patients had good tolerance to concurrent chemoradiotherapy, and both OS and PFS were higher than chemotherapy alone. The possible mechanisms for radiotherapy to prolong the survival of metastatic disease may be improve local control rate under the premise of effective systemic therapy, produce certain cytokines that further inhibit the proliferation and metastasis of tumor cells. However, the phase II study was presented only in an abstract format, and the value of radiotherapy in the treatment of metastatic EC has not yet been fully evaluated. Thus, it was initially confirmed that the survival benefits of radiotherapy in metastatic EC based on this retrospective and propensity-matched study, which laid the foundation for the following clinical studies.

However, the addition of radiotherapy to chemotherapy showed no survival benefits, which was significantly inconsistent with the study mentioned above. The reason for the discrepancy between the two conclusions may be that the specific combination modalities of chemotherapy and radiotherapy such as concurrent or sequential therapy were unknown in this study which might influence survival, and that chemotherapy plays a leading role in the treatment of metastatic EC rather than radiotherapy.

The liver is the most common metastatic site of metastatic EC (47.7% of all enrolled patients), there are only few case reports that have reported the local treatment of hepatic metastasis with good clinical efficacy (15,16), while this study showed no survival benefits in matched patients with liver metastasis when radiotherapy was applied. As for patients with more than 3 metastatic sites, radiotherapy did not improve survival, indicating the limitations of local treatment for patients with multiple metastases.

The majority of studies found that the prognosis of patients with ADC is much better than the patients with SCC (17,18), however, these studies included all clinical stages of EC, but there was no significant prognostic difference between ADC and SCC in matched patients with advanced stage, and patients of both two types showed survival benefits from radiotherapy. Besides, the multivariable Cox regression analysis demonstrated that age, sex, insurance status, differentiation, chemotherapy were also important prognostic factors for metastatic EC.

There were few limitations in this study. Firstly, The SEER database does not provide information about the sites (both primary and metastatic sites) and dose of radiation therapy. Thus, there were no data of treatment response of radiotherapy, which might affect the results of analysis. Secondly, information relating to comorbidities and performance status was not available in the SEER database, which may influence the treatment approach, resulting in selective bias. Thirdly, the impact of chemotherapy regimens was unknown as no data was available in the SEER database. Last, the combination modalities of radiation and chemotherapy had not been shown in the SEER database, which is worthy of further investigations to maximize the survival benefits of radiotherapy in metastatic EC.


Conclusions

This large population-based study demonstrated that radiotherapy could improve the survival of patients with metastatic EC, which provides a line of evidence to guide the current treatment. Further randomized studies are warranted to assess the value of radiotherapy in the management of metastatic EC.


Acknowledgments

Funding: None.


Footnote

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2019.06.15). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). Data were retrieved from SEER database. The study was approved by the Ethics Committee of Renmin Hospital of Wuhan University. Informed consent was waived.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Domper Arnal MJ, Ferrandez Arenas A, Lanas Arbeloa A. Esophageal cancer: Risk factors, screening and endoscopic treatment in Western and Eastern countries. World J Gastroenterol 2015;21:7933-43. [Crossref] [PubMed]
  2. Pennathur A, Gibson MK, Jobe BA, et al. Oesophageal carcinoma. Lancet 2013;381:400-12. [Crossref] [PubMed]
  3. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin 2017;67:7-30. [Crossref] [PubMed]
  4. Cooper JS, Guo MD, Herskovic A, et al. Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85-01). Radiation Therapy Oncology Group. JAMA 1999;281:1623-7. [Crossref] [PubMed]
  5. Li QQ, Liu MZ, Hu YH, et al. Definitive concomitant chemoradiotherapy with docetaxel and cisplatin in squamous esophageal carcinoma. Dis Esophagus 2010;23:253-9. [Crossref] [PubMed]
  6. Kleinberg L, Forastiere AA. Chemoradiation in the management of esophageal cancer. J Clin Oncol 2007;25:4110-7. [Crossref] [PubMed]
  7. Conroy T, Galais MP, Raoul JL, et al. Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 2014;15:305-14. [Crossref] [PubMed]
  8. Shridhar R, Almhanna K, Meredith KL, et al. Radiation therapy and esophageal cancer. Cancer Control 2013;20:97-110. [Crossref] [PubMed]
  9. Chun SG, Skinner HD, Minsky BD. Radiation Therapy for Locally Advanced Esophageal Cancer. Surg Oncol Clin N Am 2017;26:257-76. [Crossref] [PubMed]
  10. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin 2016;66:115-32. [Crossref] [PubMed]
  11. Torre LA, Siegel RL, Ward EM, et al. Global Cancer Incidence and Mortality Rates and Trends--An Update. Cancer Epidemiol Biomarkers Prev 2016;25:16-27. [Crossref] [PubMed]
  12. Sykes AJ, Burt PA, Slevin NJ, et al. Radical radiotherapy for carcinoma of the oesophagus: an effective alternative to surgery. Radiother Oncol 1998;48:15-21. [Crossref] [PubMed]
  13. Wu SG, Xie WH, Zhang ZQ, et al. Surgery Combined with Radiotherapy Improved Survival in Metastatic Esophageal Cancer in a Surveillance Epidemiology and End Results Population-based Study. Sci Rep 2016;6:28280. [Crossref] [PubMed]
  14. Li T, Lv J, Li F, et al. Prospective Randomized Phase 2 Study of Concurrent Chemoradiation Therapy (CCRT) Versus Chemotherapy Alone in Stage IV Esophageal Squamous Cell Carcinoma (ESCC). Int J Radiat Oncol Biol Phys 2016;96:S1. [Crossref]
  15. Huddy JR, Thomas RL, Worthington TR, et al. Liver metastases from esophageal carcinoma: is there a role for surgical resection? Dis Esophagus 2015;28:483-7. [Crossref] [PubMed]
  16. Muroi H, Nakajima M, Satomura H, et al. Effectiveness of proton beam therapy on liver metastases of esophageal cancer: report of a case. Int Surg 2015;100:180-4. [Crossref] [PubMed]
  17. Gertler R, Stein HJ, Langer R, et al. Long-term outcome of 2920 patients with cancers of the esophagus and esophagogastric junction: evaluation of the New Union Internationale Contre le Cancer/American Joint Cancer Committee staging system. Ann Surg 2011;253:689-98. [Crossref] [PubMed]
  18. Siewert JR, Ott K. Are squamous and adenocarcinomas of the esophagus the same disease? Semin Radiat Oncol 2007;17:38-44. [Crossref] [PubMed]
Cite this article as: Li X, Zhang H, Jia X, Xu L, Liu H, Chen L, Song Q, Hui Z. Survival benefit of radiotherapy in metastatic esophageal cancer: a population-based study. Transl Cancer Res 2019;8(4):1074-1085. doi: 10.21037/tcr.2019.06.15

Download Citation