A SEER prognostic nomogram analysis for extensive-stage small cell lung cancer: comparing the immune checkpoint inhibitors (ICIs) and non-ICIs eras
Original Article

A SEER prognostic nomogram analysis for extensive-stage small cell lung cancer: comparing the immune checkpoint inhibitors (ICIs) and non-ICIs eras

Xiaomei Jiang1,2, Zhongqi Wang1,2, Xiufeng Zhang2,3, Liangyun Xie1, Shanshan Ji1, Qinqin Song1, Jing Dong1, Ye Jin4, Shuangli Li1, Zhi Zhang1, Xuemei Zhang2,3

1Department of Medical Oncology, Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China; 2College of Life Sciences, North China University of Science and Technology, Tangshan, China; 3School of Public Health, North China University of Science and Technology, Tangshan, China; 4College of Clinical Medicine, North China University of Science and Technology, Tangshan, China

Contributions: (I) Conception and design: Xuemei Zhang, X Jiang, Z Zhang; (II) Administrative support: Xuemei Zhang, Z Zhang; (III) Provision of study materials or patients: X Jiang, L Xie, S Ji, Q Song; (IV) Collection and assembly of data: J Dong, Y Jin, S Li; (V) Data analysis and interpretation: X Jiang, Z Wang, Xiufeng Zhang; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Xuemei Zhang, PhD. College of Life Sciences, North China University of Science and Technology, 21 Bohai Road, Caofeidian Xincheng, Tangshan 063210, China; School of Public Health, North China University of Science and Technology, Tangshan, China. Email: zhangxuemei@ncst.edu.cn; Zhi Zhang, MD. Department of Medical Oncology, Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, No. 27, Wenhua Road, Lubei District, Tangshan 063000, China. Email: zhi1969@163.com.

Background: Extensive-stage small cell lung cancer (ES-SCLC) has a poor prognosis, and the advent of immune checkpoint inhibitors (ICIs) has introduced new possibilities for treatment. This study aimed to develop a novel nomogram to predict survival in ES-SCLC patients and identify key prognostic factors by comparing the ICIs era with the non-ICIs era.

Methods: Data were extracted from the surveillance epidemiology and end results (SEER) database, including patients diagnosed with ES-SCLC between 2015 and 2020. After using propensity score matching (PSM), patients were categorized into the ICIs era group and the non-ICIs era group (1:1 matching). Univariable and multivariable Cox regression analyses identified significant prognostic factors for overall survival (OS) and cancer-specific survival (CSS), which were used to develop nomograms for predicting 12-, 24-, and 35-month survival rates. Kaplan-Meier analysis, Cox proportional hazards models, and subgroup analyses were conducted to evaluate OS and CSS. A nomogram was developed using multivariate Cox regression analysis and validated with calibration and decision curve analyses.

Results: Immunotherapy significantly improved OS and CSS, with ICIs era patients showing a 14% reduction in OS risk [hazard ratio (HR) = 0.86, 95% confidence interval (CI): 0.82–0.90, P<0.001] and a 15% reduction in CSS risk (HR =0.85, 95% CI: 0.81–0.90, P<0.001) compared to the non-ICIs era. Significant prognostic factors included chemotherapy, liver metastasis, age, gender, marital status, income, N stage, and metastasis sites. Subgroup analyses indicated that the combination of immunotherapy and chemotherapy was particularly beneficial for female patients without liver metastases. The nomograms demonstrated higher predictive accuracy than the tumor, node, metastasis (TNM) staging system, with a Harrell’s C-index of 0.691 compared to 0.514 for TNM staging. The receiver operating characteristic (ROC) curves and decision curve analysis (DCA) further confirmed the superior performance and clinical utility of the nomograms.

Conclusions: Immunotherapy significantly enhances survival outcomes in ES-SCLC patients. The developed nomograms provide a more accurate and clinically useful tool for predicting patient survival compared to the traditional TNM staging system. These findings support the integration of immunotherapy and the use of nomograms in clinical decision-making for ES-SCLC patients.

Keywords: Extensive-stage small cell lung cancer (ES-SCLC); immune checkpoint inhibitors (ICIs); prognostic nomogram; propensity score matching (PSM); Surveillance Epidemiology and End Results database (SEER database)


Submitted Nov 15, 2024. Accepted for publication Mar 21, 2025. Published online May 28, 2025.

doi: 10.21037/jtd-2024-1981


Highlight box

Key findings

• The nomogram developed in this study provides a more accurate and practical clinical tool for predicting patient survival than the traditional tumor, node, metastasis (TNM) staging system, demonstrating that immune checkpoint inhibitors (ICIs) significantly improve overall survival (OS) and cancer-specific survival (CSS) in extensive-stage small cell lung cancer (ES-SCLC) patients.

What is known and what is new?

• Chemotherapy is the primary treatment strategy for patients with ES-SCLC. The need for combination immunotherapy in this specific patient population is controversial, and there are no accurate models to predict the prognosis of patients treated with immunotherapy.

• We determined the survival benefit of immunotherapy in patients with ES-SCLC by comparing the ICIs and non-ICIs eras and provided a valuable tool for obtaining personalized survival estimates and more individualized treatment strategies.

What is the implication, and what should change now?

• Individual factors in patients with ES-SCLC influence their treatment strategies. The nomogram may help guide clinicians to supplement ES-SCLC patients with immunotherapy to improve the prognosis of these patients.


Introduction

Small cell lung cancer (SCLC) is a highly aggressive neuroendocrine tumor characterized by a high proliferation rate, early metastatic spread and poor prognosis (1,2). It accounts for approximately 13% to 15% of all lung cancers (3). The most utilized staging system, the Veterans Administration Lung Cancer Study Group (VALCSG), classifies SCLC into a limited and extensive stage SCLC (4,5). Extensive-stage SCLC is defined as any disease that exceeds the limited stage’s confines and is not contained within the radiation field (6). Approximately 70% of patients are diagnosed at this extensive stage, of which the historical 5-year overall survival (OS) rate is less than 7% (3,7).

For the past 30 years, the standard of first-line treatment for extensive-stage small cell lung cancer (ES-SCLC) has consisted of four to six cycles of etoposide combined with platinum agents (8). In recent years, immune checkpoint inhibitors (ICIs) have emerged as a promising class of anticancer agents. By blocking immune checkpoints and restoring immune cell function, ICIs have shown significant anti-tumor effects and have been approved for a wide range of malignancies including SCLC (9-11).

The phase III IMpower-133 and CASPIAN trials demonstrated that the anti-programmed death-ligand 1 (PD-L1) checkpoint blockers durvalumab and atezolizumab significantly prolonged OS, altering the treatment paradigm for ES-SCLC (12-15). Specifically, the IMpower133 phase III pilot trial demonstrated that atezolizumab combined with chemotherapy significantly improved the median progression-free survival (PFS) (5.2 vs. 4.3 months, P=0.017) and the median OS (12.3 vs. 10.3 months, P<0.007) compared to chemotherapy alone, reducing the risk of disease progression and death by 23% and 30%, respectively (12). The CASPIAN phase III trial further confirmed that durvalumab combined with chemotherapy significantly extended median OS and reduced the risk of death by 27% (14).

Despite the potential benefits of immunotherapy, its impact on survival outcomes in different patient subgroups remains unclear. Understanding these effects is crucial for optimizing treatment strategies and personalizing patient care. Several studies identified clinical features and prognostic factors associated with survival in patients with ES-SCLC and created nomogram models (16-18). However, comprehensive prognostic models that integrate these factors with immunotherapy outcomes are still lacking. This study aims to address this gap by evaluating the effects of immunotherapy on OS and cancer-specific survival (CSS) in ES-SCLC patients across different subgroups using the surveillance epidemiology and end results (SEER) database. We utilized propensity score matching (PSM) to minimize selection bias and conducted univariable and multivariable Cox regression analysis to identify significant prognostic factors. Furthermore, we developed and validated nomograms for predicting 12-, 24-, and 35-month survival rates, comparing their predictive accuracy with the traditional tumor, node, metastasis (TNM) staging system. We present this article in accordance with the TRIPOD reporting checklist (available at https://jtd.amegroups.com/article/view/10.21037/jtd-2024-1981/rc).


Methods

Data source

The SEER program, supported by the National Cancer Institute (NCI) in the USA, collects data on patient demographics, initial tumor location, morphology, diagnosis stage, initial cancer therapy, and vital status follow-up. The SEER data (https://seer.cancer.gov/, accessed Feb 27, 2024) used in this study are publicly available, so informed patient consent is not required. The study was conducted in accordance with the Declaration of Helsinki and its subsequent amendments. For this study, we utilized the SEER database of incidence-SEER 17 Registries Custom Data with additional treatment fields from the November 2022 Sub (2000–2020), using SEER*Stat 8.4.2 software.

Patient selection

We selected patients diagnosed with SCLC as a first primary malignancy between 2015 and 2020. Exclusion Criteria: primary site: no lung and bronchus; age <18 years; no pathologically diagnosed SCLC; no one primary tumor; no distant metastatic/no Staging IV. And other exclusions: diagnosis by autopsy/death certificates; incomplete demographic/clinicopathology/therapeutic information; missing cause-specific death classification or survival status. Patients with extensive SCLC were then further screened according to National Comprehensive Cancer Network (NCCN) and VALCSG guidelines (18). Considering the IMpower133 trial and the US Food and Drug Administration’s approval of ICIs use, the period from 2018 to 2020 was defined as the “ICIs era” and the period from 2015 to 2017 as the “non-ICIs era”. Comparing the “ICIs era” and “non-ICIs era”, we used PSM (1:1), identifying 4,062 ES-SCLC patients in each era. The patient screening flow chart is shown in Figure 1.

Figure 1 Flowchart of the patients screening process. SCLC, small cell lung cancer; ES-SCLC, extensive-stage small cell lung cancer; ICIs, immune checkpoint inhibitors.

Variables collected

In this study, we evaluated the following variables: sex (male or female); race (White, Black, or other); age (≤65, 66–77, ≥78 years) determined by X-Tile software (ver.3.6.1); marital status (married, unmarried); income (<$35,000, $35,000–$74,999, $75,000+); rural-urban continuum (metropolitan, non-metropolitan); primary site [C34.0-main bronchus, C34.1-upper lobe, lung, C34.2-middle lobe, lung, C34.3-lower lobe, lung, C34.8-overlapping lesion of lung, C34.9-lung, not otherwise specified (NOS)]; grade (I–II, III–IV, or unknown); T staging (T0, T1, T2, T3, T4, Tx); N staging (N0, N1, N2, N3, Nx); M staging (M1a, M1b, M1c); tumor size (≤146, >146 mm, unknown); histologic type (small cell carcinoma, NOS; oat cell carcinoma; small cell carcinoma, fusiform cell; small cell carcinoma, intermediate cell; combined small cell carcinoma); metastasis (bone, brain, liver, or lung); visceral and pleural invasion (VPI) (yes or no); primary site surgery (yes or no); radiotherapy (yes or no); chemotherapy (yes or no); vital status (dead, alive); cause-specific death classification (cancer-related, other); survival months.

PSM

To reduce selection bias, PSM was applied using the “Matchit” R package with 1:1 matching, nearest-neighbor matching method, and a caliper width of 0.02. The following variables were used for matching: age, sex, race, marital status, income, area, primary site, T stage, N stage, grade, tumor size, brain metastasis, bone metastasis, liver metastasis, lung metastasis, surgery (primary site), radiotherapy, chemotherapy, VPI.

The nomogram and clinical prediction

A nomogram was developed to predict prognosis based on multivariable analysis. The model’s performance was evaluated using the concordance index (C-index), with a value ranging from 0 to 1. Time-dependent receiver operating characteristic (ROC) curves were used to predict OS, and the corresponding area under the curve (AUC) was calculated to demonstrate the model’s discrimination ability. Calibration curves were plotted to assess the consistency between predicted and actual probabilities. Decision curve analysis (DCA) was generated to assess the clinical benefits. All analyses were performed using R platform (ver. 4.3.2).

Statistical analysis

The baseline demographic characteristics of patients were compared using a two-tailed Chi-squared test for categorical variables with a P value less than 0.05. Kaplan-Meier (KM) analysis was used to calculate OS and cause-specific survival, with log-rank tests. Univariate and multivariate Cox proportional hazards models was used to determine prognostic variable influence on OS and CSS.


Results

Patient characteristics and PSM

A total of 9,907 ES-SCLC patients were diagnosed between 2015 to 2020 for this study, with 4,825 in the ICIs era group and 5,082 in the non-ICIs era group. X-plots were used to determine the optimal thresholds for age and tumor size, which were finally determined to be 65 and 77 years and 146 mm (Figure 2). Using these optimal thresholds as a benchmark, patients were stratified according to age and tumor size. The basic characteristics of the patients are outlined in Table 1, patients with ES-SCLC were slightly more likely to be male than female (52.07% vs. 42.93%) and were mostly Caucasian, accounting for 80.9% of the total. Metastases in patients were most frequently found in the liver, followed by bone, brain, and lung. The significant differences between the two groups of patients included income, grading, TN staging, bone metastases, brain metastases, lung metastases, and VPI. In order to minimize the effect of confounding variables, a 1:1 PSM strategy was employed, resulting in the creation of a matched cohort of 4,062 patients in each of the “ICIs” era and “non-ICIs” era groups. The baseline characteristics of the two cohorts showed a high degree of consistency. The standardized mean difference (SMD) between the matched cohorts was consistently below 0.1, indicating significant homogeneity in the distribution of scores (Figure 3).

Figure 2 Defining the optimal cutoffs of age and tumor size via X-tile analysis. The black dot indicates that optimal cutoff values of age/tumor size have been identified (A,B). A histogram of age/tumor size (C,D). Kaplan-Meier curve of age/tumor size (E,F). Optimal cutoff values of age were 65 and 77 years. Optimal cutoff values of tumor size were 146 mm.

Table 1

Baseline characteristics of patients before and after propensity score matching

Characteristic Before PSM (N=9,907) After PSM (N=8,124)
ICIs era
(N=4,825)
Non-ICIs era (N=5,082) P value SMD ICIs era
(N=4,062)
Non-ICIs era (N=4,062) P value SMD
Sex 0.84 0.004 >0.99 <0.001
   Female 2,307 (47.8) 2,441 (48.0) 1,946 (47.9) 1,945 (47.9)
   Male 2,518 (52.2) 2,641 (52.0) 2,116 (52.1) 2,117 (52.1)
Age (years) 0.94 0.007 0.94 0.008
   ≤65 2,182 (45.2) 2,284 (44.9) 1,842 (45.3) 1,846 (45.4)
   ≥78 627 (13.0) 657 (12.9) 521 (12.8) 530 (13.0)
   66–77 2,016 (41.8) 2,141 (42.1) 1,699 (41.8) 1,686 (41.5)
Race 0.44 0.026 0.95 0.007
   Black 424 (8.8) 435 (8.6) 336 (8.3) 338 (8.3)
   Other 267 (5.5) 254 (5.0) 206 (5.1) 212 (5.2)
   White 4,134 (85.7) 4,393 (86.4) 3,520 (86.7) 3,512 (86.5)
Marital 0.22 0.025 0.89 0.003
   Married 2,341 (48.5) 2,530 (49.8) 1,972 (48.5) 1,979 (48.7)
   Unmarried 2,484 (51.5) 2,552 (50.2) 2,090 (51.5) 2,083 (51.3)
Income ($) <0.001 0.273 0.99 0.004
   35,000–74,999 2,627 (54.4) 3,391 (66.7) 2,493 (61.4) 2,487 (61.2)
   ≥75,000 2,098 (43.5) 1,547 (30.4) 1,471 (36.2) 1,478 (36.4)
   <35,000 100 (2.1) 144 (2.8) 98 (2.4) 97 (2.4)
Area 0.68 0.009 >0.99 0.001
   Metropolitan 3,846 (79.7) 4,033 (79.4) 3,187 (78.5) 3,188 (78.5)
   Non-metropolitan 979 (20.3) 1,049 (20.6) 875 (21.5) 874 (21.5)
Primary site 0.65 0.037 0.93 0.026
   Main bronchus 538 (11.2) 572 (11.3) 452 (11.1) 468 (11.5)
   Upper lobe, lung 2,209 (45.8) 2,363 (46.5) 1,891 (46.6) 1,849 (45.5)
   Middle lobe, lung 178 (3.7) 182 (3.6) 150 (3.7) 149 (3.9)
   Lower lobe, lung 1,031 (21.4) 1,058 (20.8) 860 (21.2) 861 (21.2)
   Overlapping lesion of lung 90 (1.9) 74 (1.5) 63 (1.6) 69 (1.7)
   Lung, NOS 779 (16.1) 833 (16.4) 646 (15.9) 666 (16.4)
Grade <0.001 0.123 0.97 0.017
   I 4 (0.1) 3 (0.1) 3 (0.1) 2 (0.0)
   II 5 (0.1) 13 (0.3) 5 (0.1) 6 (0.1)
   III 402 (8.3) 443 (8.7) 338 (8.3) 348 (8.6)
   IV 334 (6.9) 514 (10.1) 324 (8.0) 332 (8.2)
   Unknown 4,080 (84.6) 4,109 (80.9) 3,392 (83.5) 3,374 (83.1)
T stage <0.001 0.218 0.92 0.027
   T0 68 (1.4) 48 (0.9) 51 (1.3) 45 (1.1)
   T1 499 (10.3) 478 (9.4) 422 (10.4) 424 (10.4)
   T2 774 (16.0) 1,079 (21.2) 731 (18.0) 760 (18.7)
   T3 726 (15.0) 1,014 (20.0) 679 (16.7) 694 (17.1)
   T4 1,986 (41.2) 1,833 (36.1) 1,603 (39.5) 1,574 (38.7)
   TX 772 (16.0) 630 (12.4) 576 (14.2) 565 (13.9)
N stage <0.001 0.14 0.97 0.016
   N0 621 (12.9) 570 (11.2) 490 (12.1) 487 (12.0)
   N1 334 (6.9) 340 (6.7) 282 (6.9) 277 (6.8)
   N2 2,137 (44.3) 2,558 (50.3) 1,919 (47.2) 1,908 (47.0)
   N3 1,384 (28.7) 1,359 (26.7) 1,139 (28.0) 1,166 (28.7)
   NX 349 (7.2) 255 (5.0) 232 (5.7) 224 (5.5)
Tumor size (mm) 0.92 0.008 0.86 0.012
   >146 41 (0.8) 47 (0.9) 30 (0.7) 34 (0.8)
   ≤146 3,656 (75.8) 3,846 (75.7) 3,095 (76.2) 3,084 (75.9)
   Unknown 1,128 (23.4) 1,189 (23.4) 937 (23.1) 944 (23.2)
Bone met <0.001 0.075 0.88 0.011
   No 2,865 (59.4) 3,073 (60.5) 2,434 (59.9) 2,446 (60.2)
   Unknown 43 (0.9) 84 (1.7) 40 (1.0) 36 (0.9)
   Yes 1,917 (39.7) 1,925 (37.9) 1,588 (39.1) 1,580 (38.9)
Brain met 0.002 0.07 0.87 0.012
   No 3,288 (68.1) 3,530 (69.5) 2,805 (69.1) 2,824 (69.5)
   Unknown 50 (1.0) 87 (1.7) 44 (1.1) 41 (1.0)
   Yes 1,487 (30.8) 1,465 (28.8) 1,213 (29.9) 1,197 (29.5)
Liver met 0.21 0.036 0.93 0.008
   No 2,604 (54.0) 2,729 (53.7) 2,186 (53.8) 2,183 (53.7)
   Unknown 43 (0.9) 64 (1.3) 35 (0.9) 32 (0.8)
   Yes 2,178 (45.1) 2,289 (45.0) 1,841 (45.3) 1,847 (45.5)
Lung met <0.001 0.112 0.77 0.016
   No 3,939 (81.6) 3,932 (77.4) 3,249 (80.0) 3,253 (80.1)
   Unknown 68 (1.4) 116 (2.3) 66 (1.6) 58 (1.4)
   Yes 818 (17.0) 1,034 (20.3) 747 (18.4) 751 (18.5)
Surgery primary site 0.25 0.025 >0.99 <0.001
   No 4,767 (98.8) 5,034 (99.1) 4,021 (99.0) 4,021 (99.0)
   Yes 58 (1.2) 48 (0.9) 41 (1.0) 41 (1.0)
Radiotherapy 0.22 0.025 0.81 0.006
   No 2,744 (56.9) 2,827 (55.6) 2,280 (56.1) 2,292 (56.4)
   Yes 2,081 (43.1) 2,255 (44.4) 1,782 (43.9) 1,770 (43.6)
Chemotherapy 0.82 0.005 0.81 0.006
   No/unknown 1,053 (21.8) 1,120 (22.0) 890 (21.9) 880 (21.7)
   Yes 3,772 (78.2) 3,962 (78.0) 3,172 (78.1) 3,182 (78.3)
VPI <0.001 0.077 0.57 0.015
   No 4,710 (97.6) 5,014 (98.7) 4,008 (98.7) 4,001 (98.5)
   Yes 115 (2.4) 68 (1.3) 54 (1.3) 61 (1.5)

Data are presented as n (%). ICIs, immune checkpoint inhibitors; NOS, not otherwise specified; PSM, propensity score matching; SMD, standardized mean difference; VPI, visceral and pleural invasion.

Figure 3 Propensity score matching between the ICIs group and non-ICIs group. The SMD between the ICIs group and non-ICIs group (A). Propensity score (B) and distribution of propensity scores (C). ICIs, immune checkpoint inhibitors; SMD, standardized mean difference.

OS and CSS analysis before and after PSM

Prior to PSM, a total of 9,907 patients were included, and the median OS or CSS time was 7 months in both the ICIs era and non-ICIs era groups. The ICI era group showed a better prognosis, with OS rates at 6-, 12-, 24-, and 33-month of 52.23% vs. 51.32%, 24.79% vs. 20.20%, and 9.50% vs. 3.36%, and 5.87% vs. 0.58%, respectively. Similarly, the CSS rates at 6, 12, 24, and 33-month were 54.69% vs. 53.51%, 26.78% vs. 22.14%, and 10.97% vs. 4.07%, and 7.07% vs. 0.84%, respectively (Table 2). After PSM, the results exhibited no statistically significant disparities from those before matching. The median OS or CSS time for both groups remained around 7 months. However, the ICI era group continued to show a better prognosis, with 6-, 12-, 24-, and 33-month OS rates of 52.48% vs. 51.49%, 24.51% vs. 20.09%, 9.13% vs. 3.42%, and 5.74% vs. 0.56%, respectively. The corresponding CSS rates were 55.09% vs. 53.72%, 26.52% vs. 22.02%, 10.55% vs. 4.04%, and 6.96% vs. 0.84%, respectively.

Table 2

Survival rates of patients stratified by treatment method

Variables Original cohort (n=9,907) Matched cohort (n=8,124)
ICIs era group, n=4,825 Non-ICIs era group, n=5,082 P value ICIs era group, n=4,062 Non-ICIs era group, n=4,062 P value
OS (%) (95% CI) <0.001 <0.001
   6-month 52.23 (50.79–53.72) 51.32 (49.96–52.72) 52.48 (50.90–54.10) 51.49 (49.97–53.05)
   12-month 24.79 (23.46–26.21) 20.20 (19.13–21.34) 24.51 (23.06–26.04) 20.09 (18.89–21.37)
   24-month 9.50 (8.41–10.74) 3.36 (2.90–3.91) 9.13 (7.98–10.44) 3.42 (2.90–4.03)
   33-month 5.87 (4.48–7.70) 0.58 (0.40– 0.84) 5.74 (4.27–7.71) 0.56 (0.37–0.86)
   Median OS (range) 7 (7–7) 7 (6–7) 7 (7–7) 7(6–7)
CSS (%) (95% CI) <0.001 <0.001
   6-month 54.69 (53.23–56.18) 53.51 (52.14–54.92) 55.09 (53.50–56.72) 53.72 (52.19–55.29)
   12-month 26.78 (25.38–28.27) 22.14 (20.99–23.35) 26.52 (24.99–28.13) 22.02 (20.74–23.38)
   24-month 10.97 (9.74–12.35) 4.07 (3.53–4.69) 10.55 (9.26–12.02) 4.04 (3.44–4.75)
   33-month 7.07 (5.48–9.14) 0.84 (0.60–1.19) 6.96 (5.27–9.19) 0.84 (0.57–1.24)
   Median CSS (range) 7 (7–8) 7 (7–7) 7 (7–8) 7 (7–7)

CI, confidence interval; CSS, cancer-specific survival; ICIs, immune checkpoint inhibitors; OS, overall survival.

KM analysis compared OS and CSS in the ICI era versus the non-ICI era groups before and after PSM. Patients in the ICIs group had significantly improved OS and CSS compared with the non-ICIs group (all P<0.001) (Figure 4A,4B). After propensity matching, immunotherapy significantly prolonged OS and CSS in ES-SCLC patients (all P<0.001) (Figure 4C,4D).

Figure 4 Survival differences in each era (non-ICIs and ICIs). Kaplan-Meier curves for OS and CSS of patients before PSM (A,B) and after PSM (C,D). CI, confidence interval; CSS, cancer-specific survival; HR, hazard ratio; ICIs, immune checkpoint inhibitors; OS, overall survival; PSM, propensity score matching.

Cox regression analysis of OS and CSS

Univariable and multivariable Cox regression analyses of OS (Table 3) and CSS (Table 4) were conducted in patients with ES-SCLC, spanning three years in the ICI era and three years in the pre-ICI era. The univariable Cox regression analysis revealed that group, sex, age, marital status, income, N stage, bone metastasis, brain metastasis, liver metastasis, lung metastasis, radiotherapy, and chemotherapy were significantly correlated with OS and CSS (P<0.01), which were included in further multivariate analyses.

Table 3

Univariable and multivariable cox regression analyses for overall survival of patients

Characteristic N Event N Univariable Multivariable
HR 95% CI P value HR 95% CI P value
Group
   Non-ICIs era 4,062 4,030
   ICIs era 4,062 3,049 0.85 0.81, 0.89 <0.001 0.86 0.82, 0.90 <0.001
Age (years)
   ≤65 3,688 3,140
   ≥78 1,051 942 1.69 1.57, 1.82 <0.001 1.47 1.36, 1.58 <0.001
   66–77 3,385 2,997 1.22 1.16, 1.28 <0.001 1.18 1.12, 1.24 <0.001
Sex
   Male 4,233 3,708
   Female 3,891 3,371 0.91 0.87, 0.96 <0.001 0.92 0.87, 0.96 <0.001
Race
   Black 674 593
   White 7,032 6,137 0.99 0.91, 1.08 0.86
   Other 418 349 0.95 0.83, 1.08 0.41
Marital
   Unmarried 4,173 3,658
   Married 3,951 3,421 0.90 0.86, 0.95 <0.001 0.88 0.84, 0.93 <0.001
Income ($)
   ≥75,000 2,949 2,520
   35,000–74,999 4,980 4,395 1.10 1.05, 1.16 <0.001 1.13 1.07, 1.18 <0.001
   <35,000 195 164 1.06 0.90, 1.24 0.49 1.15 0.98, 1.35 0.09
Area
   Metropolitan 6,375 5,559
   Non-metropolitan 1,749 1,520 1.04 0.99, 1.11 0.13
Primary site
   Main bronchus 920 803
   Lower lobe, lung 1,721 1,509 1.03 0.95, 1.13 0.46 0.95 0.87, 1.03 0.20
   Middle lobe, lung 299 251 0.90 0.78, 1.04 0.15 0.85 0.74, 0.98 0.03
   Upper lobe, lung 3,740 3,234 0.96 0.89, 1.04 0.34 0.93 0.86, 1.00 0.06
   Overlapping lesion of lung 132 112 1.04 0.85, 1.27 0.69 1.00 0.82, 1.22 0.99
   Lung, NOS 1,312 1,170 1.13 1.03, 1.24 0.008 1.01 0.93, 1.11 0.76
Grade
   I 5 4
   II 11 10 3.33 1.04, 10.62 0.04
   III 686 602 2.61 0.98, 6.98 0.06
   IV 656 548 2.45 0.92, 6.55 0.07
   Unknown 6,766 5,915 2.62 0.98, 6.97 0.06
T stage
   T0 96 89
   T1 846 743 0.93 0.75, 1.16 0.54
   T2 1,491 1,300 0.93 0.75, 1.16 0.54
   T3 1,373 1,197 1.03 0.83, 1.28 0.80
   T4 3,177 2,744 1.00 0.81, 1.23 0.97
   TX 1,141 1,006 1.11 0.89, 1.38 0.36
N stage
   N0 977 852
   N1 559 480 0.92 0.82, 1.02 0.12 1.00 0.89, 1.12 0.96
   N2 3,827 3,347 1.07 0.99, 1.15 0.08 1.23 1.14, 1.33 <0.001
   N3 2,305 1,991 1.04 0.96, 1.13 0.36 1.27 1.17, 1.38 <0.001
   NX 456 409 1.27 1.13, 1.43 <0.001 1.13 1.00, 1.27 0.06
Tumor size (mm)
   >146 64 56
   ≤146 6,179 5,360 0.85 0.66, 1.11 0.24
   Unknown 1,881 1,663 0.94 0.72, 1.23 0.66
Bone met
   No 4,880 4,211
   Yes 3,168 2,804 1.14 1.09, 1.19 <0.001 1.19 1.13, 1.25 <0.001
   Unknown 76 64 1.37 1.07, 1.75 0.01 1.02 0.76, 1.35 0.92
Brain met
   No 5,629 4,881
   Yes 2,410 2,123 1.08 1.03, 1.14 0.003 1.36 1.28, 1.44 <0.001
   Unknown 85 75 1.49 1.19, 1.88 <0.001 1.10 0.84, 1.44 0.48
Liver met
   No 4,369 3,700
   Yes 3,688 3,321 1.38 1.32, 1.45 <0.001 1.42 1.35, 1.49 <0.001
   Unknown 67 58 1.42 1.09, 1.83 0.009 0.87 0.63, 1.20 0.41
Lung met
   No 6,502 5,641
   Yes 1,498 1,331 1.16 1.09, 1.23 <0.001 1.10 1.04, 1.17 0.002
   Unknown 124 107 1.24 1.02, 1.50 0.03 0.91 0.74, 1.12 0.39
Surgery primary site
   No 8,042 7,011
   Yes 82 68 0.73 0.58, 0.93 0.01
Radiotherapy
   No 4,572 4,053
   Yes 3,552 3,026 0.68 0.64, 0.71 <0.001 0.77 0.73, 0.81 <0.001
Chemotherapy
   No/unknown 1,770 1,693
   Yes 6,354 5,386 0.30 0.28, 0.32 <0.001 0.31 0.29, 0.33 <0.001
VPI
   No 8,009 6,974
   Yes 115 105 0.92 0.76, 1.11 0.39

–, the reference group. CI, confidence interval; HR, hazard ratio; ICIs, immune checkpoint inhibitors; NOS, not otherwise specified; VPI, visceral and pleural invasion.

Table 4

Univariable and multivariable cox regression analyses for cancer-specific survival of patients

Characteristic N Event N Univariable Multivariable
HR 95% CI P value HR 95% CI P value
Group
   Non-ICIs era 4,062 3,813
   ICIs era 4,062 2,859 0.84 0.80, 0.88 <0.001 0.85 0.81, 0.90 <0.001
Age (years)
   ≤65 3,688 2,963
   ≥78 1,051 879 1.67 1.55, 1.80 <0.001 1.46 1.35, 1.58 <0.001
   66–77 3,385 2,830 1.22 1.16, 1.29 <0.001 1.18 1.12, 1.24 <0.001
Sex
   Male 4,233 3,483
   Female 3,891 3,189 0.92 0.88, 0.96 <0.001 0.93 0.88, 0.97 0.002
Race
   Black 674 551
   White 7,032 5,793 1.01 0.92, 1.10 0.84
   Other 418 328 0.96 0.83, 1.10 0.53
Marital
   Unmarried 4,173 3,414
   Married 3,951 3,258 0.92 0.88, 0.97 0.001 0.90 0.86, 0.95 <0.001
Income ($)
   ≥75,000 2,949 2,370
   35,000–74,999 4,980 4,151 1.11 1.05, 1.16 <0.001 1.13 1.08, 1.19 <0.001
   <35,000 195 151 1.03 0.88, 1.22 0.69 1.13 0.95, 1.33 0.16
Area
   Metropolitan 6,375 5,253
   Non-metropolitan 1,749 1,419 1.03 0.97, 1.09 0.29
Primary Site
   Main bronchus 920 760
   Lower lobe, lung 1,721 1,415 1.02 0.94, 1.12 0.61
   Middle lobe, lung 299 237 0.90 0.78, 1.04 0.15
   Upper lobe, lung 3,740 3,047 0.96 0.89, 1.04 0.30
   Overlapping lesion of lung 132 111 1.09 0.90, 1.33 0.38
   Lung, NOS 1,312 1,102 1.12 1.03, 1.23 0.01
Grade
   I 5 4
   II 11 9 2.99 0.92, 9.70 0.07
   III 686 570 2.48 0.93, 6.64 0.07
   IV 656 519 2.33 0.87, 6.23 0.09
   Unknown 6,766 5,570 2.47 0.93, 6.59 0.07
T stage
   T0 96 83
   T1 846 690 0.93 0.74, 1.17 0.53
   T2 1,491 1,222 0.94 0.75, 1.18 0.60
   T3 1,373 1,130 1.04 0.83, 1.30 0.72
   T4 3,177 2,605 1.02 0.82, 1.26 0.89
   TX 1,141 942 1.11 0.89, 1.39 0.35
N stage
   N0 977 797
   N1 559 452 0.92 0.82, 1.04 0.17 1.00 0.89, 1.13 0.97
   N2 3,827 3,178 1.09 1.01, 1.18 0.04 1.25 1.15, 1.35 <0.001
   N3 2,305 1,866 1.04 0.96, 1.13 0.34 1.28 1.17, 1.39 <0.001
   NX 456 379 1.26 1.12, 1.43 <0.001 1.14 1.00, 1.29 0.05
Tumor size (mm)
   >146 64 52
   ≤146 6,179 5,042 0.86 0.66, 1.13 0.29
   Unknown 1,881 1,578 0.96 0.73, 1.27 0.78
Bone met
   No 4,880 3,951
   Yes 3,168 2,661 1.15 1.10, 1.21 <0.001 1.21 1.15, 1.27 <0.001
   Unknown 76 60 1.37 1.06, 1.77 0.02 1.04 0.77, 1.39 0.81
Liver met
   No 4,369 3,487
   Yes 3,688 3,131 1.39 1.32, 1.46 <0.001 1.42 1.35, 1.50 <0.001
   Unknown 67 54 1.40 1.07, 1.83 0.01 0.90 0.65, 1.26 0.55
Brain met
   No 5,629 4,581
   Yes 2,410 2,024 1.10 1.04, 1.16 <0.001 1.37 1.30, 1.46 <0.001
   Unknown 85 67 1.41 1.11, 1.80 0.005 1.04 0.79, 1.38 0.77
Lung met
   No 6,502 5,324
   Yes 1,498 1,248 1.15 1.08, 1.22 <0.001 1.10 1.03, 1.17 0.003
   Unknown 124 100 1.22 1.00, 1.49 0.05 0.93 0.75, 1.15 0.49
Surgery primary site
   No 8,042 6,608
   Yes 82 64 0.73 0.57, 0.94 0.01
Radiotherapy
   No 4,572 3,793
   Yes 3,552 2,879 0.69 0.65, 0.72 <0.001 0.78 0.74, 0.82 <0.001
Chemotherapy
   No/unknown 1,770 1,602
   Yes 6,354 5,070 0.30 0.28, 0.31 <0.001 0.30 0.29, 0.32 <0.001
VPI
   No 8,009 6,572
   Yes 115 100 0.93 0.76, 1.13 0.47

–, the reference group. CI, confidence interval; HR, hazard ratio; ICIs, immune checkpoint inhibitors; NOS, not otherwise specified; VPI, visceral and pleural invasion.

The multivariable Cox regression analysis indicated that, age, gender, marital status, income, N stage, bone metastasis, brain metastasis, liver metastasis, lung metastasis, radiotherapy, and chemotherapy served as independent prognostic factors for both CSS and OS in ICIs era group (Figure 5).

Figure 5 Forest plots displaying prognostic factors by the multivariable Cox regression analysis. Analysis prognostic factors for OS (A) and CSS (B). CSS, cancer-specific survival; CI, confidence interval; HR, hazard ratio; ICIs, immune checkpoint inhibitors; NOS, not otherwise specified; OS, overall survival.

Age over 65 years at diagnosis showed a negative impact on OS [hazard ratio (HR) =1.18, 95% confidence interval (CI): 1.12–1.24, P<0.001]. Other negative prognostic factors included income less than $75,000 (HR =1.13, 95% CI: 1.07–1.18, P<0.001), N2 stage (HR =1.23, 95% CI: 1.14–1.33, P<0.001), N3 stage (HR =1.27, 95% CI: 1.17–1.38, P<0.001), bone metastasis (HR =1.19, 95% CI: 1.13–1.25, P<0.001), brain metastasis (HR =1.36; 95% CI: 1.28–1.44, P<0.001), liver metastasis (HR =1.42, 95% CI: 1.35–1.49, P<0.001), and lung metastasis (HR =1.10, 95% CI: 1.04–1.17, P=0.002). Conversely, ICIs era group (HR =0.86, 95% CI: 0.82–0.90, P<0.001), being married (HR =0.88, 95% CI: 0.84–0.93, P<0.001), female (HR =0.92, 95% CI: 0.87–0.96, P<0.001), radiotherapy (HR =0.77, 95% CI: 0.73–0.81, P<0.001), and chemotherapy (HR =0.31, 95% CI: 0.29–0.33, P<0.001) were associated with favorable OS outcomes.

The ICIs era group demonstrated a significant survival advantage compared to the non-ICIs era group, with a 14% reduction in OS time (HR =0.86, 95% CI: 0.82–0.90, P<0.001) and a 15% reduction in CSS time (HR =0.85, 95% CI: 0.81–0.90, P<0.001). These findings underscore the substantial protective of immunotherapy on CSS and OS in patients with ES-SCLC. Furthermore, we analyzed the interval from diagnosis to treatment for patients with ES-SCLC in each subgroup (Tables S1,S2). This interval represents the time from definitive diagnosis to the initiation of primary treatment and serves as a key indicator of both healthcare system efficiency and patient adherence to treatment. Notably, shorter intervals may reflect a higher level of patient motivation and adherence to treatment, which could positively influence prognosis in ES-SCLC patients.

Effects of immunotherapy on ES-SCLC patients in different subgroups

To determine the effects of immunotherapy on ES-SCLC patients in different subgroups, we conducted subgroup analyses and interaction tests. Figure 6A illustrates that immunotherapy serves as a protective factor for OS or CSS in ES-SCLC patients in ICIs era. The effect on OS was consistent across all 16 subgroups (all P for interaction >0.05), with the exceptions being income, liver metastasis, and chemotherapy (all P for interaction <0.01). The effect of immunotherapy on CSS was same as that of OS, indicating robustness of the overall effect. Notable differences were observed in the interaction of the four subgroup variables (gender, income, liver metastases, and chemotherapy) in the ICI era (all P for interaction <0.05) (Figure 6B). Regarding metastatic patterns, our analysis revealed that single-site metastases were similar between the ICIs and non-ICIs eras, with hepatic metastases being the most prevalent (45.1% vs. 45.0%) and bone metastases affecting a substantial proportion of patients (39.7% vs. 37.9%) (Table S3). Additionally, brain metastases were slightly more frequent in the ICIs era (30.8% vs. 28.8%), while lung metastases were more common in the non-ICIs era (20.3% vs. 17.0%). However, when analyzing multiple metastatic sites, notable differences emerged. Specifically, the co-occurrence of bone and brain metastases was significantly higher in the ICIs era (15.4%) than in the non-ICIs era (8.6%), suggesting a potential shift in metastatic distribution with immunotherapy use. Similarly, other metastatic combinations, such as brain and liver metastases (10.0% vs. 8.8%) and bone, brain, and liver metastases (5.5% vs. 4.7%), were slightly more prevalent in the ICIs era. These findings provide important insights into metastatic trends in the era of immunotherapy and their potential prognostic implications. At the same time, we found further KM analysis revealed that the combination of immunotherapy and chemotherapy may be especially beneficial for patients with ES-SCLC without liver metastases (Figure S1). In the subgroup analysis, OS or CSS benefits were observed across all subgroups in ICIs era compared non-ICIs era, except for the following subgroups: non-white or non-black race, main bronchus and middle lobe lung sites, grade I or II, T0 or Tx stage, N0 or Nx lymph node, unknown metastasis, primary site surgery, and VPI involvement.

Figure 6 Subgroup analysis and interaction tests of ICIs era or non-ICIs era group after PSM. Subgroup analysis for OS (A) and CSS (B). CI, confidence interval; CSS, cancer-specific survival; HR, hazard ratio; ICIs, immune checkpoint inhibitors; NOS, not otherwise specified; OS, overall survival; PSM, propensity score matching; VPI, visceral and pleural invasion.

Construction and validation of nomograms for prognostic prediction

Twelve prognostic indicators identified through univariable and multivariable Cox regression analyses were used to develop the final nomograms. Figure 7A presents the nomograms for predicting 12-, 24-, and 35-month OS rates. Chemotherapy was identified as the strongest prognostic factor, followed by liver metastasis and age. Each level of these predictors is assigned a specific score in the nomogram. By summing the scores for each predictor, total scores are obtained, which then allow for the estimation of 12-, 24-, and 35-month survival probabilities for individuals based on a vertical line from the total points row.

Figure 7 Nomograms for predicting prognosis and the calibration curves for predicting 1-year OS in ES-SCLC patients. The nomogram model (A). Nomogram calibration curves (B). X-axis: the OS probability predicted by the nomogram; Y-axis: the actual OS probability. A plot along the 45-degree dotted line indicates a perfect calibration model, where the predicted probabilities and actual outcomes are identical. ES-SCLC, extensive-stage small cell lung cancer; ICIs, immune checkpoint inhibitors; OS, overall survival.

The Harrell’s C-index for the established nomogram was 0.691 (95% CI: 0.683–0.699, P<0.001), significantly higher than that for the TNM staging system [odds ratio (OR) =0.514, 95% CI: 0.529–0.565, P<0.001]. Figure 7B presents the 1,000-sample bootstrapped calibration plot for predicting 1-year OS, revealing good predictive accuracy of the nomogram. To evaluate the performance of the nomogram, receiver operating characteristic curves were plotted. Comparison analysis showed that the area under the AUC value of OS nomogram is higher than that of the TNM staging (0.697 vs. 0.514) (Figure 8A,8B). The DCAs of OS compared the net benefits of the nomograms with the TNM staging system. Figure 8C demonstrates that the 1- and 2-year outcomes of the nomograms surpass those of the TNM staging system in addressing various risk factors for death in the matched cohort. This indicates that this new model offers better clinical utility and enhances practical decision-making.

Figure 8 ROC curves of nomogram and TNM stage for predicting OS rates in ES-SCLC patients and DCA curves compares the net benefits of the novel nomogram and TNM staging system. ROC curves of nomogram (A). ROC curves of TNM stage (B). DCA curves of 1- and 2-year OS compares the net benefits of the novel nomogram and TNM staging system (C). AUC, area under the curve; DCA, decision curve analysis; ES-SCLC, extensive-stage small cell lung cancer; OS, overall survival; ROC, receiver operating characteristic; TNM, tumor-node-metastasis.

Discussion

ES-SCLC is a challenging malignancy with a poor prognosis and limited treatment options. Traditionally, the first-line therapy for ES-SCLC has been a combination of etoposide with platinum. Although this regimen achieves high response rates, the responses are often short-lived, with a median OS of about 10 months, and frequent relapse (19,20). The introduction of ICIs has revolutionized the SCLC treatment, with drugs like nivolumab, pembrolizumab, atezolizumab, and durvalumab significantly improving clinical outcomes (9,10,12,21). Phase III clinical trials, such as IMpower133, CASPIAN, have demonstrated that combination ICIs and chemotherapy can substantially extend OS in ES-SCLC patients (12-15). This study provides comprehensive insights into the impact of immunotherapy on OS and CSS in patients with ES-SCLC. Using SEER database, we compared outcomes in the ICIs era with those in the non-ICI era and analyzed various prognostic factors through PSM, univariable and multivariable Cox regression analyses, and nomogram development. Our results demonstrated that immunotherapy significantly improved OS and CSS in ES-SCLC patients. Specifically, patients in the ICIs era showed a 14% reduction in OS risk and a 15% reduction in CSS risk compared to those in the non-ICIs era. These findings align with the outcomes of pivotal clinical trials such as IMpower-133 and CASPIAN, which established the efficacy of PD-L1 inhibitors in combination with chemotherapy.

Significant prognostic factors identified in our study included chemotherapy, liver metastasis, age, gender, marital status, income, N stage, and metastasis sites. Among these, chemotherapy emerged as the strongest prognostic factor, underscoring its critical role in the treatment regimen for ES-SCLC. Our subgroup analyses further revealed that the combination of immunotherapy and chemotherapy is particularly beneficial for female patients without liver metastases, highlighting the need for personalized treatment strategies. The advent of the ICI era has seen consistent results from phase III trials (e.g., IMpower133, CASPIAN, KEYNOTE-604, CAPSTONE-1, ASTRUM-005, RATIONALE-312, ETER701) demonstrating that the integration of ICIs with platinum-based chemotherapy has revolutionized first-line treatment approach in ES-SCLC patients (12-15,22-26). Notably, the benefits of ICIs have been observed to be consistent regardless of whether cisplatin or carboplatin is used. The addition of ICIs to platinum-based chemotherapy has been observed to improve patient quality of life compared with chemotherapy alone (20). Despite several challenges, including the need for individualized therapy and the lack of biomarkers, immunotherapy has great potential to improve survival and quality of life in patients with SCLC. Therefore, the importance of individualized therapy is growing, and the integration of multiple clinical factors of the patient is essential to tailor the treatment regimen. Additionally, Survival analysis revealed that liver metastases had the worst OS and CSS (both P<0.05), which was consistent with previous findings (27).

The nomograms developed in this study demonstrated superior predictive accuracy compared to the traditional TNM staging system. In line with the report which (28) showed that the Harrell’s C-index for our nomogram was 0.691, significantly higher than the 0.514 for the TNM system. The ROC curves and DCAs also confirmed the enhanced clinical utility of our nomograms. These tools provide clinicians with a robust method for predicting individualized patient survival, facilitating more informed and tailored treatment decisions in ICIs era.

Despite these promising findings, there are several limitations in this study. Firstly, although eastern cooperative oncology group (ECOG) score performance status is a well-established predictor of survival in ES-SCLC (29,30), it is not available in the SEER database, preventing its incorporation into our prognostic nomogram. The absence of this variable may introduce potential bias, as performance status plays a crucial role in treatment decisions and patient outcomes.

Secondly, paraneoplastic syndromes and blood biomarkers, such as neutrophil-to-lymphocyte ratio (NLR), lactate dehydrogenase (LDH), and pro-gastrin-releasing peptide (ProGRP), have been recognized as important prognostic factors in ES-SCLC (31-33). However, since these variables are not recorded in the SEER database, we were unable to include them in our analysis, which may have limited the predictive power of our nomogram. Additionally, paraneoplastic syndromes can influence both treatment response and survival outcomes. Future studies integrating real-world clinical data with these biomarkers would be valuable in refining and validating prognostic models for ES-SCLC.

Thirdly, while treatment adherence is a critical factor influencing prognosis, the SEER database does not provide specific data on the number of treatment cycles completed by patients. Instead, we analyzed the interval from diagnosis to treatment initiation as a proxy measure of adherence. Shorter intervals may indicate greater patient motivation and access to care, potentially leading to improved survival outcomes. However, this metric does not fully capture adherence throughout the treatment course. Future studies incorporating detailed treatment completion data would provide a more comprehensive understanding of adherence and its prognostic implications.

Finally, the SEER database lacks detailed records of prior medical and treatment histories, preventing us from identifying patients who initially received chemo-radiotherapy for limited-stage disease before progressing to extensive-stage disease. This limitation restricts our ability to evaluate the impact of previous treatment exposure on metastatic patterns and survival outcomes. Future studies incorporating real-world clinical data with comprehensive treatment histories would be valuable in further elucidating the influence of prior therapies on metastatic behavior and response to immunotherapy.

Despite these limitations, our study provides valuable insights into prognostic factors in ES-SCLC, and our nomogram demonstrated superior predictive accuracy compared to the TNM staging system. Future studies incorporating detailed clinical variables, biomarker data, and treatment adherence metrics will further improve prognostic modeling and clinical decision-making in ES-SCLC (34-36).


Conclusions

In conclusion, this study underscores the significant survival benefits of immunotherapy in ES-SCLC patients and highlights the importance of incorporating prognostic factors into treatment planning. The development of a novel nomogram that integrates traditional prognostic factors and immunotherapy provides a superior tool for predicting individualized patient survival. This nomogram can enhance clinical decision-making and facilitate personalized treatment planning, thereby improving patient outcomes.


Acknowledgments

The authors would like to express our appreciation for the efforts of the Surveillance, Epidemiology, and End Results (SEER) Program cancer registries in the creation of the SEER database.


Footnote

Reporting Checklist: The authors have completed the TRIPOD reporting checklist. Available at https://jtd.amegroups.com/article/view/10.21037/jtd-2024-1981/rc

Peer Review File: Available at https://jtd.amegroups.com/article/view/10.21037/jtd-2024-1981/prf

Funding: This work was supported by the Foundation for Talents by Tangshan Human Resources and Social Security Bureau (No. A202110007), Hebei Province 2024 Annual Medical Science Research Project Plan (No. 20241895) and the Clinical Research Special Funding Fund of Wu Jieping Medical Foundation (No. 320.6750.2021-02-100).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jtd.amegroups.com/article/view/10.21037/jtd-2024-1981/coif). 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. The study was conducted in accordance with the Declaration of Helsinki and its subsequent amendments.

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. Megyesfalvi Z, Gay CM, Popper H, et al. Clinical insights into small cell lung cancer: Tumor heterogeneity, diagnosis, therapy, and future directions. CA Cancer J Clin 2023;73:620-52. [Crossref] [PubMed]
  2. Rudin CM, Brambilla E, Faivre-Finn C, et al. Small-cell lung cancer. Nat Rev Dis Primers 2021;7:3. [Crossref] [PubMed]
  3. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12-49. [Crossref] [PubMed]
  4. Ganti AKP, Loo BW, Bassetti M, et al. Small Cell Lung Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021;19:1441-64. [Crossref] [PubMed]
  5. Lee JH, Saxena A, Giaccone G. Advancements in small cell lung cancer. Semin Cancer Biol 2023;93:123-8. [Crossref] [PubMed]
  6. Giunta EF, Addeo A, Rizzo A, et al. First-Line Treatment for Advanced SCLC: What Is Left Behind and Beyond Chemoimmunotherapy. Front Med (Lausanne) 2022;9:924853. [Crossref] [PubMed]
  7. Gazdar AF, Bunn PA, Minna JD. Small-cell lung cancer: what we know, what we need to know and the path forward. Nat Rev Cancer 2017;17:725-37. [Crossref] [PubMed]
  8. Waqar SN, Morgensztern D. Treatment advances in small cell lung cancer (SCLC). Pharmacol Ther 2017;180:16-23. [Crossref] [PubMed]
  9. Zugazagoitia J, Osma H, Baena J, et al. Facts and Hopes on Cancer Immunotherapy for Small Cell Lung Cancer. Clin Cancer Res 2024;30:2872-83. [Crossref] [PubMed]
  10. Liu M, Qiu G, Guan W, et al. Induction chemotherapy followed by camrelizumab plus apatinib and chemotherapy as first-line treatment for extensive-stage small-cell lung cancer: a multicenter, single-arm trial. Signal Transduct Target Ther 2025;10:65. [Crossref] [PubMed]
  11. Han X, Guo J, Li L, et al. Sintilimab combined with anlotinib and chemotherapy as second-line or later therapy in extensive-stage small cell lung cancer: a phase II clinical trial. Signal Transduct Target Ther 2024;9:241. [Crossref] [PubMed]
  12. Horn L, Mansfield AS, Szczęsna A, et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N Engl J Med 2018;379:2220-9. [Crossref] [PubMed]
  13. Goldman JW, Dvorkin M, Chen Y, et al. Durvalumab, with or without tremelimumab, plus platinum-etoposide versus platinum-etoposide alone in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): updated results from a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2021;22:51-65. [Crossref] [PubMed]
  14. Paz-Ares L, Dvorkin M, Chen Y, et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet 2019;394:1929-39. [Crossref] [PubMed]
  15. Liu SV, Reck M, Mansfield AS, et al. Updated Overall Survival and PD-L1 Subgroup Analysis of Patients With Extensive-Stage Small-Cell Lung Cancer Treated With Atezolizumab, Carboplatin, and Etoposide (IMpower133). J Clin Oncol 2021;39:619-30. [Crossref] [PubMed]
  16. Zhong J, Zheng Q, An T, et al. Nomogram to predict cause-specific mortality in extensive-stage small cell lung cancer: A competing risk analysis. Thorac Cancer 2019;10:1788-97. [Crossref] [PubMed]
  17. Li J, Liu F, Yu H, et al. Different distant metastasis patterns based on tumor size could be found in extensive-stage small cell lung cancer patients: a large, population-based SEER study. PeerJ 2019;7:e8163. [Crossref] [PubMed]
  18. Zou J, Guo S, Xiong MT, et al. Ageing as key factor for distant metastasis patterns and prognosis in patients with extensive-stage Small Cell Lung Cancer. J Cancer 2021;12:1575-82. [Crossref] [PubMed]
  19. Wang S, Zimmermann S, Parikh K, et al. Current Diagnosis and Management of Small-Cell Lung Cancer. Mayo Clin Proc 2019;94:1599-622. [Crossref] [PubMed]
  20. Gomez-Randulfe I, Leporati R, Gupta B, et al. Recent advances and future strategies in first-line treatment of ES-SCLC. Eur J Cancer 2024;200:113581. [Crossref] [PubMed]
  21. Owonikoko TK, Park K, Govindan R, et al. Nivolumab and Ipilimumab as Maintenance Therapy in Extensive-Disease Small-Cell Lung Cancer: CheckMate 451. J Clin Oncol 2021;39:1349-59. [Crossref] [PubMed]
  22. Rudin CM, Awad MM, Navarro A, et al. Pembrolizumab or Placebo Plus Etoposide and Platinum as First-Line Therapy for Extensive-Stage Small-Cell Lung Cancer: Randomized, Double-Blind, Phase III KEYNOTE-604 Study. J Clin Oncol 2020;38:2369-79. [Crossref] [PubMed]
  23. Wang J, Zhou C, Yao W, et al. Adebrelimab or placebo plus carboplatin and etoposide as first-line treatment for extensive-stage small-cell lung cancer (CAPSTONE-1): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:739-47. [Crossref] [PubMed]
  24. Cheng Y, Han L, Wu L, et al. Effect of First-Line Serplulimab vs Placebo Added to Chemotherapy on Survival in Patients With Extensive-Stage Small Cell Lung Cancer: The ASTRUM-005 Randomized Clinical Trial. JAMA 2022;328:1223-32. [Crossref] [PubMed]
  25. Cheng Y, Fan Y, Zhao Y, et al. Tislelizumab Plus Platinum and Etoposide Versus Placebo Plus Platinum and Etoposide as First-Line Treatment for Extensive-Stage SCLC (RATIONALE-312): A Multicenter, Double-Blind, Placebo-Controlled, Randomized, Phase 3 Clinical Trial. J Thorac Oncol 2024;19:1073-85. [Crossref] [PubMed]
  26. Cheng Y, Chen J, Zhang W, et al. Benmelstobart, anlotinib and chemotherapy in extensive-stage small-cell lung cancer: a randomized phase 3 trial. Nat Med 2024;30:2967-76. [Crossref] [PubMed]
  27. Cai H, Wang H, Li Z, et al. The prognostic analysis of different metastatic patterns in extensive-stage small-cell lung cancer patients: a large population-based study. Future Oncol 2018;14:1397-407. [Crossref] [PubMed]
  28. Liu M, Zhang P, Wang S, et al. Comparation between novel online models and the AJCC 8th TNM staging system in predicting cancer-specific and overall survival of small cell lung cancer. Front Endocrinol (Lausanne) 2023;14:1132915. [Crossref] [PubMed]
  29. Agarwal M, Liu A, Langlais BT, et al. Chemoimmunotherapy as the First-Line Treatment for Patients With Extensive-Stage Small-Cell Lung Cancer and an ECOG Performance Status 2 or 3. Clin Lung Cancer 2023;24:591-7. [Crossref] [PubMed]
  30. Rittberg R, Green S, Aquin T, et al. Effect of Hospitalization During First Chemotherapy and Performance Status on Small-cell Lung Cancer Outcomes. Clin Lung Cancer 2020;21:e388-404. [Crossref] [PubMed]
  31. Soomro Z, Youssef M, Yust-Katz S, et al. Paraneoplastic syndromes in small cell lung cancer. J Thorac Dis 2020;12:6253-63. [Crossref] [PubMed]
  32. Iams WT, Shiuan E, Meador CB, et al. Improved Prognosis and Increased Tumor-Infiltrating Lymphocytes in Patients Who Have SCLC With Neurologic Paraneoplastic Syndromes. J Thorac Oncol 2019;14:1970-81. [Crossref] [PubMed]
  33. Sebastian M, Koschade S, Stratmann JA. SCLC, Paraneoplastic Syndromes, and the Immune System. J Thorac Oncol 2019;14:1878-80. [Crossref] [PubMed]
  34. Xie M, Vuko M, Rodriguez-Canales J, et al. Molecular classification and biomarkers of outcome with immunotherapy in extensive-stage small-cell lung cancer: analyses of the CASPIAN phase 3 study. Mol Cancer 2024;23:115. [Crossref] [PubMed]
  35. Cui Y, Chen Y, Zhao P, et al. Peripheral NK cells identified as the predictor of response in extensive-stage small cell lung cancer patients treated with first-line immunotherapy plus chemotherapy. Clin Transl Oncol 2024;26:2522-30. [Crossref] [PubMed]
  36. Liu Q, Zhang J, Guo C, et al. Proteogenomic characterization of small cell lung cancer identifies biological insights and subtype-specific therapeutic strategies. Cell 2024;187:184-203.e28. [Crossref] [PubMed]
Cite this article as: Jiang X, Wang Z, Zhang X, Xie L, Ji S, Song Q, Dong J, Jin Y, Li S, Zhang Z, Zhang X. A SEER prognostic nomogram analysis for extensive-stage small cell lung cancer: comparing the immune checkpoint inhibitors (ICIs) and non-ICIs eras. J Thorac Dis 2025;17(5):2866-2887. doi: 10.21037/jtd-2024-1981

Download Citation