Efficacy evaluation of surgery combined with chemotherapy for stage IIIA small cell lung cancer patients: a retrospective analysis

This article has an erratum available at: http://dx.doi.org/10.21037/tlcr-2022-9 the article has been update on 2022-11-28 at here.

Original Article

Efficacy evaluation of surgery combined with chemotherapy for stage IIIA small cell lung cancer patients: a retrospective analysis

Dongliang Bian1,2#, Siming Jiang2#, Yicheng Xiong1,2#, Mengfan Qi1,2, Jiawei Wu1,2, Alfredo Addeo3, Yoshikane Yamauchi4, Farkhad Manapov5, Wolfram C. M. Dempke6, Jacopo Vannucci7, Alessandro Di Federico8, Xiaoxiong Xu2, Linsong Chen2

1Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China; 2Department of Thoracic Surgery, Tongji University Affiliated Shanghai Pulmonary Hospital, Shanghai, China; 3Oncology Department, University Hospital of Geneva, Geneva, Switzerland; 4Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan; 5Department of Radiation Oncology, University Hospital Ludwig Maximilian University of Munich (LMU), Munich, Germany; 6Department of Haematology and Oncology, University Medical School Munich, Munich, Germany; 7Thoracic Surgery Unit, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy; 8Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy

Contributions: (I) Conception and design: D Bian; (II) Administrative support: L Chen, X Xu; (III) Provision of study materials or patients: L Chen; (IV) Collection and assembly of data: S Jiang, M Qi; (V) Data analysis and interpretation: D Bian, Y Xiong; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Linsong Chen. Department of Thoracic Surgery, Tongji University Affiliated Shanghai Pulmonary Hospital, 507# Zhengmin Road, Yangpu District, Shanghai 200433, China. Email: chenlinsong1223@126.com.

Background: The efficacy of surgery in combination of chemotherapy for stage IIIA small cell lung cancer (IIIA-SCLC) is controversial. The aim of the present study was to analyze the efficacy of surgery combined with chemotherapy, especially in the setting of neoadjuvant chemotherapy (NAC) followed by surgery for IIIA-SCLC.

Methods: Between 2004 and 2015, we reviewed 2,199 chemotherapy-treated stage IIIA (N1/2) SCLC cases in the Surveillance, Epidemiology, and End Results (SEER) database, and 32 NAC + intentional radical resection-treated, centrally-located IIIA-SCLC cases at Shanghai Pulmonary Hospital (SPH). Outcomes were compared between surgically and non-surgically treated patients from the SEER database after propensity score matching (PSM), and comparing lobectomy/bi-lobectomy and pneumonectomy patients from SPH. Prognostic factors were evaluated by Kaplan-Meier method and the Cox proportional hazards regression model.

Results: There was significantly higher overall survival (OS) in surgically treated IIIA-SCLC patients (OS, 44.8 vs. 21.2 months, P=0.048), and similar efficacy was observed between sub-lobectomy and lobectomy/bi-lobectomy patients (OS: 55.6 vs. 30.3 months, P=0.167) in SEER database. At SPH, significantly higher OS was associated with T1 stage (before NAC: T1 vs. T2–4, 48.7 vs. 32.2 months, P=0.025; after NAC: T1 vs. T2–4, 42.7 vs. 21.3 months, P=0.048). Female sex [hazard ratio (HR): 0.078, P=0.009], T1 stage (HR: 13.048, P=0.026), and pneumonectomy (HR: 0.095, P=0.009) were independent prognostic factors for IIIA-SCLC patients who received NAC + intentional radical resection.

Conclusions: For stage IIIA SCLC patients, complete resection combined with chemotherapy might improve the prognosis than patients without surgery. Post-NAC lobectomy was not found to be superior to sub-lobectomy, while pneumonectomy was considered suitable for central-type IIIA-SCLC patients after NAC treatment.

Keywords: Small cell lung cancer (SCLC); limited disease; neoadjuvant chemotherapy (NAC); surgery


Submitted May 13, 2022. Accepted for publication Aug 10, 2022.

doi: 10.21037/tlcr-22-545


Introduction

Small cell lung cancer (SCLC) is one of the most aggressive type of lung cancer accounting for approximately 15% of thoracic malignancies (1-3). Although early diagnosis and the therapeutic regimens for SCLC have improved in the past 4 decades, improvement in overall survival (OS) remains rather poor (4-7). Previously published studies have demonstrated that patients with untreated limited and extensive SCLC (LD-SCLC and ED-SCLC) have an OS of about 6 and 2 months, respectively, and more than 80% of the patients undergoing standard therapies died within 2 years after diagnosis (8,9). For early-stage SCLC, particularly with no regional lymph node involvement, surgery combined with chemotherapy was the standard therapeutic regimen, and was found to significantly prolong OS (10-12). At the time of diagnosis, more than one third of patients who developed hilar and/or mediastinal lymph node (N1 and N2) involvement, which was defined as locally advanced SCLC (LA-SCLC) (13-15). Because of high recurrence and low OS rates for surgical treated LA-SCLC, surgery has been excluded from potential multimodality regimens since the 1990s (16-18). Current multimodality regimens based on surgery, chemotherapy, targeted therapy, immune checkpoint inhibitors (ICIs), and anti-angiogenic inhibitors (AAIs) have offered positive outcomes in the treatment of stage IIIA SCLC (IIIA-SCLC) disease, according to the TNM staging system for lung cancer (19-24). However, the therapeutic regimen for IIIA-SCLC is still controversial. A number of randomized clinical trials (RCT) on resectable stage I–IIIA SCLC have been carried out to analyze the efficacy of surgery combined with induction therapies (18,25).

In the present article, we reviewed chemotherapy-treated IIIA-SCLC patients in the Surveillance, Epidemiology, and End Results (SEER) database, and neoadjuvant chemotherapy (NAC) + intentional radical resection-treated, central-type IIIA-SCLC at Tongji University affiliated with Shanghai Pulmonary Hospital (SPH).

The aim of the present study was to analyze the efficacy of intentional radical resection in combination with chemotherapy for IIIA-SCLC patients, and to explore the potentially optimal surgical procedures and adjuvant therapeutic regimens. We present the following article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-545/rc).


Methods

Subjects

From January 2004 to December 2015, we reviewed patients diagnosed with IIIA-SCLC in the SEER database. The TNM stage of all patients was reclassified according to the American Joint Committee on Cancer TNM staging system (8th edition). The patient-selection process is outlined in Figure 1. The exclusion criteria were: (I) diagnosis prior to January 2004 or after December 2015; (II) age >80 years; (III) no cytologic/pathologic confirmation in the diagnostic information; (IV) no regional lymph node involvement; (V) missing clinical and/or follow-up information; (VI) patients not treated with chemotherapy; and (VII) patients with previous history of cancer.

Figure 1 Flowchart of the Surveillance, Epidemiology, and End Results database research selection process. IIIA-SCLC, stage IIIA small cell lung cancer.

Measures and procedures

Patient characteristics, including age at diagnosis, sex, differentiated grade, tumor laterality, tumor stage (T stage), lymph node stage (N stage), therapeutic regimens, and outcomes were collected. Patients were divided into the surgical group and non-surgical group. To eliminate the possibility of selection bias between the 2 groups, 1:1 propensity score matching (PSM) was performed by using the caliper match algorithm with a width of 0.1 of the standard deviation of the logit for the propensity score to evaluate the efficacy of surgical treatment for IIIA-SCLC patients with regional lymph node involvement (N1 or N2 according to TNM 8th edition). Surgically treated patients were separated into the lobectomy or bi-lobectomy group (LG), sub-lobectomy group (S-LG), and pneumonectomy group (PG) to compare the outcomes.

Between January 2004 and December 2015, 32 consecutive NAC plus radical resection-treated, central-type stage IIIA (N2) SCLC patients were reviewed at SPH. All patients in N2 stage were pathological confirmed. Postoperative follow-up was achieved by telephone contact or outpatient visit. The deadline for updating survival information in this study was June 30, 2017. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by Shanghai Pulmonary Hospital ethics committee (No. K18-066), and informed consent was taken from all the patients.

Statistics

Statistical analyses were performed by SPSS software (version 26.0; IBM, Armonk, NY, USA). PSM was performed by R version 3.6.2 software (http://www.r-project.org/). Variables of patients’ characteristics were compared by Student’s t-test, χ2-test, and Fisher’s exact test. Kaplan-Meier method and log-rank test were used to compare outcomes. OS was calculated from the time of diagnosis to death. Recurrence-free survival was calculated from the time of surgery to disease relapse (SCLC relapse was confirmed by cytology and/or pathology). Independent predictive factors were calculated using the Cox proportional hazards regression model. Two-tailed P<0.05 was considered statistically significant.


Results

Characteristics of SEER patients

A total of 2,199 patients in the SEER database who had received chemotherapy were included (Figure 1). A total of 114 and 2,085 patients received surgical and non-surgical treatment, respectively. The proportion of patients who received radiotherapy in the surgical group was significantly higher than that in the non-surgical group (71.9% vs. 24.1%, P<0.001). No significant difference in age at diagnosis was observed between the 2 groups (64 vs. 64.4 years, P=0.907). There was no significant difference in the distribution of other variables between the 2 groups. PSM was performed between the surgical and non-surgical subgroups. The number of patients in these 2 subgroups after PSM was 108 in each (Table 1).

Table 1

Baseline characteristics of patients with chemotherapy-treated IIIA-SCLC before and after PSM in the SEER database

Variables Subtypes Unmatched PSM
Surgery No surgery P value Surgery No surgery P value
Age (years) 64.0 64.4 0.907 64.2 63.5 0.521
Sex, n (%) Male 50 (43.9) 906 (43.5) 0.932 50 (46.3) 45 (41.7) 0.583
Female 64 (56.1) 1,179 (56.5) 58 (53.7) 63 (58.3)
Location, n (%) Left 52 (45.6) 776 (37.2) 0.072 51 (47.2) 50 (46.3) 1.000
Right 62 (54.4) 1,309 (63.8) 57 (52.8) 58 (53.7)
T stage, n (%) T1 47 (41.2) 778 (37.3) 0.227 47 (43.5) 55 (50.9) 0.503
T2 55 (48.2) 1,155 (55.4) 55 (50.9) 49 (45.4)
T3–4 12 (10.5) 152 (7.3) 6 (5.6) 4 (3.7)
N stage, n (%) N1 12 (10.5) 152 (7.3) 0.200 6 (5.6) 4 (3.7) 0.436
N2 102 (89.5) 1,933 (92.7) 102 (94.4) 104 (96.3)
Radiotherapy, n (%) With 82 (71.9) 502 (24.1) <0.001 76 (70.4) 76 (70.4) 1.000
Without 32 (28.1) 1,583 (75.9) 32 (29.6) 32 (29.6)
Total 114 2,085 108 108

IIIA-SCLC, stage IIIA small cell lung cancer; PSM, propensity score matching; SEER, Surveillance, Epidemiology, and End Results database.

OS analysis of patients in the SEER database

The results of univariate survival analyses after PSM were listed in Table 2. Compared with the non-surgical subgroup, a significantly higher OS was observed in IIIA-SCLC patients who received surgery + chemotherapy (OS, surgery vs. non-surgery, 44.8 vs. 21.2 months, P=0.048) (Figure 2A). As shown in Tables 3,4, 61 and 48 patients received lobectomy/bi-lobectomy and sub-lobectomy, respectively. Compared with lobectomy, sub-lobectomy could provide similar OS in NAC treated IIIA stage SCLC (OS, sub-lobectomy vs. anatomical lobectomy, 55.6 vs. 30.3 months, P=0.167) (Figure 2B).

Table 2

Univariate analysis for OS of chemotherapy-treated IIIA-SCLC after PSM in the SEER database

Variables Subtypes OS
Rate (%) Time (month) 95% CI P value
Age (years) ≤65 31.0 37.8 28.7–47.0 0.856
>65 36.0 36.8 27.0–46.6
Sex Male 33.7 36.2 26.8–45.6 0.692
Female 33.1 37.6 28.8–46.4
Location Left side 41.6 38.0 28.7–47.3 0.373
Right side 26.1 35.8 27.3–44.3
T stage T1 37.3 42.6 31.7–53.5 0.442
T2 28.8 32.1 24.8–39.4
T3–4 40.0 15.6 11.9–19.4
N stage N1 40.0 15.6 11.9–19.4 0.419
N2 33.0 38.7 31.6–45.7
Surgery With 34.3 44.8 34.5–55.2 0.048
Without 32.4 21.2 21.5–34.9
PORT With 36.2 41.2 32.7–49.8 0.077
Without 26.6 30.6 20.0–41.3

OS, overall survival; IIIA-SCLC, stage IIIA small cell lung cancer; PSM, propensity score matching; SEER, Surveillance, Epidemiology, and End Results database; CI, confidence interval; PORT, postoperative radiotherapy.

Figure 2 OS of chemotherapy-treated IIIA-SCLC with or without surgery (SG vs. CG) (A) and different surgery types (B) according to the Surveillance, Epidemiology, and End Results database patient analysis. OS of radical resection-treated IIIA-SCLC concerning different T-stage pre-NAC (C) and different responses to NAC (D) according to the SPH patient analysis. OS, overall survival; IIIA-SCLC, stage IIIA small cell lung cancer; SG, surgery group; CG, non-surgery group; S-LG, sub-lobectomy group; LG, lobectomy/bi-lobectomy group; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; NAC, neoadjuvant chemotherapy; SPH, Shanghai Pulmonary Hospital.

Table 3

Baseline characteristics of surgery + chemotherapy-treated IIIA-SCLC in the SEER database

Variables Subtypes S-LG LG PG P value
Age (years), median (range) 64.6 (62.2–67.0) 63.6 (61.3–65.9) 62.6 (47.4–77.9) 0.807
Sex, n (%) Male 18 (37.5) 30 (49.2) 2 (40.0) 0.438
Female 30 (62.5) 31 (50.8) 3 (60.0)
Location, n (%) Left 24 (50.0) 24 (39.3) 4 (80.0) 0.175
Right 24 (50.0) 37 (60.7) 1 (20.0)
T stage, n (%) T1 24 (50.0) 23 (37.7) 0 (0.0) 0.004
T2 23 (47.9) 30 (49.2) 2 (40.0)
T3–4 1 (2.1) 8 (13.1) 3 (60.0)
N stage, n (%) N1 1 (2.1) 8 (13.1) 3 (60.0) 0.002
N2 47 (97.9) 53 (86.9) 2 (40.0)
PORT, n (%) With 36 (75.0) 44 (72.1) 2 (40.0) 0.241
Without 12 (25.0) 17 (27.9) 3 (60.0)
Total 48 61 5

IIIA-SCLC, stage IIIA small cell lung cancer; SEER, Surveillance, Epidemiology, and End Results database; S-LG, sub-lobectomy group; LG, lobectomy/bi-lobectomy group; PG, pneumonectomy group; PORT, postoperative radiotherapy.

Table 4

Univariate analysis for OS and lung CSS of surgery + chemotherapy-treated IIIA-SCLC in the SEER database

Variables Subtypes OS CSS
Rate (%) Time (month) 95% CI P value Rate (%) Time (month) 95% CI P value
Age (years) <65 39.0 51.8 36.7–66.9 0.358 39.0 51.8 36.7–66.9 0.670
≥65 32.7 37.7 25.2–50.2 40.0 41.1 27.4–54.8
Sex Male 32.0 43.1 33.7–60.4 0.946 34.0 43.9 29.7–58.1 0.784
Female 39.1 47.1 29.1–57.0 43.8 50.4 36.4–64.5
Location Left side 38.5 41.6 29.2–53.9 0.652 44.2 44.8 31.6–57.9 0.656
Right side 33.9 45.9 32.1–59.7 35.5 46.9 32.7–60.7
T stage T1 40.4 55.6 38.4–72.7 0.297 42.6 56.7 39.4–74.1 0.393
T2 29.1 32.9 23.4–42.4 34.5 34.9 24.7–45.1
T3–4 50.0 38.1 20.3–55.9 50.0 38.1 20.3–55.9
N stage N1 50.0 38.1 20.3–55.9 0.864 50.0 38.1 20.3–55.9 0.989
N2 34.3 45.8 35.2–56.5 38.2 48.1 36.9–59.2
Surgery Lobectomy 29.5 30.3 22.7–37.9 0.052 31.1 30.8 23.1–38.6 0.031
Sub-lobectomy 39.6 55.6 38.7–72.5 45.8 60.0 42.2–77.8
Pneumonectomy 80.0 60.8 41.2–80.4 80.0 60.8 41.2–80.4
PORT With 36.6 50.3 38.1–62.3 0.111 40.2 52.3 39.7–65.0 0.104
Without 34.4 37.1 19.5–54.6 37.5 41.7 22.9–60.5

OS, overall survival; CSS, cancer-specified survival; IIIA-SCLC, stage IIIA small cell lung cancer; SEER, Surveillance, Epidemiology, and End Results database; CI, confidence interval; PORT, postoperative radiotherapy.

Characteristics of patients from SPH

A total of 32 centrally-located IIIA-SCLC patients (age range, 37–76 years) received NAC at SPH, and 29 patients received 2 NAC cycles (90.6%). Etoposide + carboplatin/cisplatin (EP/EC) were given to 26 patients (81.3%). Significantly better OS was observed in patients with NAC plus intentional radical resection compared with patients who received non-surgical treatment (29.7 vs. 16.2 months). According to paired-samples Student’s t-test, the median tumor size decreased significantly after NAC treatment (maximum dimension of SCLC, pre-NAC vs. post-NAC, 43 vs. 19.4 mm, P<0.001). Compared with patients in the LG, the median tumor size after NAC was significantly larger in the PG (26.8 vs. 18.9 mm, P=0.014) (Table 5).

Table 5

Baseline characteristics of neoadjuvant chemotherapy + radical surgery-treated IIIA-SCLC in SPH patients

Variables Subtypes LG (%) PG (%) P value
Age (years) 58.0 53.0 0.424
Tumor size (mm) Pre-NAC 36.2 49.9 0.281
Post-NAC 18.9 26.8 0.014
Sex Male 11 (73.3) 15 (88.2) 0.383
Female 4 (26.7) 2 (11.8)
Smoking status Yes 6 (40.0) 7 (41.2) 0.946
No 9 (60.0) 10 (58.8)
Location Left 4 (26.7) 10 (58.8) 0.067
Right 11(73.3) 7 (41.2)
T stage (pre-NAC) T1 6 (40.0) 0 (0.0) 0.006
T2–4 9 (60.0) 17 (100.0)
T stage (post-NAC) T1 9 (60.0) 11 (64.7) 0.784
T2–4 6 (40.0) 6 (35.3)
N stage (post-NAC) N0–1 6 (40.0) 3 (17.6) 0.157
N2 9 (60.0) 14 (82.4)
Pathology Pure SCLC 11 (73.3) 11 (64.7) 0.599
Combined SCLC 4 (26.7) 6 (35.3)
ADC With 12 (80.0) 16 (94.1) 0.228
Without 3 (20.0) 1 (5.9)
ADR With 4 (26.7) 8 (47.1) 0.234
Without 11(73.3) 9 (52.9)
Recurrence With 9 (60.0) 9 (52.9) 0.688
Without 6 (40.0) 8 (47.1)
Total 15 17

IIIA-SCLC, stage IIIA small cell lung cancer; SPH, Shanghai Pulmonary Hospital database; LG, lobectomy/bi-lobectomy group; PG, pneumonectomy group; NAC, neoadjuvant chemotherapy; ADC, adjuvant chemotherapy; ADR, adjuvant radiotherapy.

SPH patients outcomes

Table 6 manifested that compared with patients with advanced T stage, significantly higher OS was observed in patients with T1 stage (before NAC: 48.7 vs. 32.2 months, P=0.025; after NAC: 42.7 vs. 21.3 months, P=0.048) (Figure 2C, Table 6). Cox proportional hazards regression model revealed that the female [female: male, hazard ratio (HR): 0.078, 95% confidence interval (CI): 0.012–0.525, P=0.009], T1 stage at diagnosis (T2–4: T1, HR: 13.048, 95% CI: 1.358–125.385, P=0.026), and pneumonectomy (pneumonectomy: lobectomy, HR: 0.095, 95% CI: 0.016–0.555, P=0.009) were independent prognostic factors for OS (Table 7).

Table 6

Univariate analysis for OS and RFS of neoadjuvant chemotherapy + radical surgery-treated IIIA-SCLC in SPH patients

Variables Subtypes RFS OS
Rate (%) Time (month) 95% CI P value Rate (%) Time (month) 95% CI P value
Age <55 years 40.0 29.7 16.1–43.4 0.632 26.7 30.4 19.6–41.2 0.640
≥55 years 47.1 38.8 21.5–56.0 35.3 38.4 24.5–51.9
Sex Male 46.2 37.6 23.5–51.7 0.890 30.8 35.3 24.7–45.9 0.693
Female 33.3 28.8 11.0–46.6 33.3 33.5 16.5–50.5
Smoking status Yes 46.2 29.8 13.8–45.8 0.672 30.8 26.7 15.1–38.3 0.353
No 42.1 37.2 22.0–52.3 31.6 39.8 27.6–52.0
Location Left side 28.6 23.2 10.3–36.0 0.109 28.6 29.2 17.3–41.0 0.367
Right side 55.6 44.6 27.6–61.7 33.3 39.3 26.9–51.7
T stage (pre-NAC) T1 50.0 36.2 15.3–57.1 0.545 66.7 48.7 35.8–61.6 0.025
T2–4 42.3 34.4 20.6–48.1 23.1 32.2 22.1–43.4
T stage (post-NAC) T1 45.5 39.7 25.1–54.2 0.325 40.9 42.7 30.6–54.8 0.048
T2–4 40.0 20.5 10.5–30.5 10.0 21.3 11.6–31.1
N stage (post-NAC) N0–1 44.4 33.9 16.3–51.5 0.625 44.4 40.7 26.2–55.2 0.166
N2 43.5 33.3 18.7–48.1 26.1 31.1 20.8–41.5
Response to NAC Downstaging 50.0 44.4 22.9–65.9 0.340 50.0 50.6 32.3–68.9 0.052
No downstaging 40.9 25.8 15.1–36.5 22.7 26.3 19.1–33.4
Pathology Pure SCLC 45.5 40.3 26.0–54.6 0.164 40.9 41.4 28.7–54.1 0.230
Combined SCLC 40.0 27.3 9.3–45.4 10.0 25.4 15.1–35.7
NAC plan EC/EP 46.2 39.8 26.1–53.5 0.161 34.6 39.4 28.8–50.0 0.062
Other 33.3 18.7 4.0–33.3 16.7 19.7 4.5–34.9
Surgery Lobectomy/bi-lobectomy 40.0 27.7 14.3–41.2 0.643 33.3 31.9 21.1–42.6 0.983
Pneumonectomy 47.1 38.5 21.5–55.4 29.4 36.7 33.7–49.8
ADC With 42.9 34.4 20.8–48.0 0.382 28.6 33.4 23.3–43.6 0.191
Without 50.0 32.8 20.7–44.8 50.0 46.1 31.6–60.7
ADR With 33.3 32.5 14.5–50.4 0.723 33.3 38.4 24.6–52.2 0.566
Without 50.0 34.5 21.9–47.1 30.0 30.3 20.2–40.3

OS, overall survival; RFS, recurrence-free survival; IIIA-SCLC, stage IIIA small cell lung cancer; SPH, Shanghai Pulmonary Hospital database; CI, confidence interval; NAC, neoadjuvant chemotherapy; ADC, adjuvant chemotherapy; ADR, adjuvant radiotherapy; EC/EP, etoposide + carboplatin/cisplatin.

Table 7

Multivariate analysis for OS and RFS of neoadjuvant chemotherapy + radical surgery-treated IIIA-SCLC in SPH patients

Variables (reference) Subtypes RFS OS
Hazard ratio 95% CI P value Hazard ratio 95% CI P value
Age, years (<55) ≥55 0.435 0.125–1.517 0.192 0.772 0.225–2.655 0.682
Sex (male) Female 0.483 0.066–3.529 0.474 0.078 0.012–0.525 0.009
Smoking status (no) Yes 0.585 0.258–11.025 0.585 0.772 0.151–3.950 0.756
Location (left side) Right side 0.179 0.046–0.704 0.014 0.543 0.151–1.945 0.348
Pre-NAC T stage (T1) T2–4 2.773 0.231–33.260 0.421 13.048 1.358–125.385 0.026
Post-NAC T stage (T1) T2–4 0.924 0.088–9.651 0.947 3.473 0.697–17.299 0.129
Post-NAC N stage (N0–1) N2 1.223 0.434–3.450 0.703 2.774 0.778–9.895 0.116
Response to NAC (DS) Non-DS 1.197 0.313–4.575 0.793 2.887 0.731–11.410 0.131
Pathology (pure SCLC) Combined SCLC 8.424 1.177–60.295 0.034 0.905 0.265–3.088 0.873
NAC plan (EC/EP) Other 3.185 0.578–17.552 0.183 4.446 0.801–24.686 0.088
Surgery (lobectomy/bi-lobectomy) Pneumonectomy 0.195 0.028–1.349 0.098 0.095 0.016–0.555 0.009
ADC (without) With 1.187 0.087–16.1805 0.898 2.831 0.322–24.9093 0.348
ADR (without) With 4.268 0.631–28.847 0.137 1.215 0.220–6.701 0.823

OS, overall survival; RFS, recurrence-free survival; IIIA-SCLC, stage IIIA small cell lung cancer; SPH, Shanghai Pulmonary Hospital database; NAC, neoadjuvant chemotherapy; DS, downstaging; ADC, adjuvant chemotherapy; ADR, adjuvant radiotherapy; CI, confidence interval.


Discussion

SCLC remains a very aggressive and lethal malignant disease. The core treatment for SCLC is chemotherapy in combination with anti-PD-L1 antibodies, for which EP/EC plus Atezolizumab/Durvalumab, particularly for LA-SCLC and ED-SCLC, is most used and significantly improved prognosis (7,22,25-29). The efficacy of surgery combined with chemotherapy for LA-SCLC remains controversial (10). In the past 3 decades, surgically treated LA-SCLC has been relatively rare, particularly SCLC with regional lymph node involvement (4-6). Our analysis of patients in the SEER database indicated that, compared with non-surgical treated LA-SCLC, a significantly higher OS was associated with surgery plus chemotherapy (median OS: 44.8 vs. 21.2 months). The results of the SPH patient analysis largely verified our findings from our SEER database analysis that NAC plus surgery is potentially an effective treatment for IIIA-SCLC.

Previously published studies have demonstrated that the strongest prognostic factor for SCLC is regional lymph node involvement (30). Significantly higher OS has been observed in patients with neoadjuvant therapy plus radical resection (19,20,31). Based on these findings, surgical treatment might not be considered as a contraindication for IIIA-SCLC, and neoadjuvant and adjuvant chemotherapy or immunotherapy should be planned to enhance therapeutic effects.

The criteria for IIIA-SCLC surgery needs to be improved. First, NAC-treated patients whose tumor size reduces to less than 3 cm and receives radical resection could have significantly better OS. Therefore, a significantly better OS could be offered through radical-resection surgery to patients who have a consistent response to NAC. Moreover, IIIA-SCLC OS could be prolonged by chemotherapy plus surgery, regardless of the status of regional lymph node involvement. These results also support the findings of a previously published study that reported that a significant difference in OS was not observed between N1 and N2 lymph node-positive patients (19). Then, compared with patients who achieved complete or partial response after NAC, SCLC patients with stable disease followed by surgery could have worse OS, although no statistical difference is observed (Figure 2D). Therefore, no matter how neoadjuvant therapeutic responses are, NAC plus surgery, including pneumonectomy, is potentially beneficial for IIIA-SCLC patients.

With the expansion of surgical indications for LA-SCLC, the investigation of what surgery was performed is also carried out. In clinical work, individualized surgical strategy for different patients is in line with the basic surgical principles. Firstly, it is understandable that N1 involvement and advanced clinical T stage LA-SCLC patients were treated by lobectomy or bi-lobectomy. They could remove disease completely and could significantly improve patients’ outcomes. Second, for IIIA-SCLC with N2 involvement and a relatively smaller tumor size, radical resection is difficult, while sub-lobectomy could meet the needs of complete resection for primary lesions and preserve the postoperative pulmonary function of patients (32). According to the results of the SEER database analysis, a better outcome of IIIA-SCLC was not observed by extended resection, especially for patients with N2 involvement. Sub-lobectomy combined with appropriate adjuvant therapies is a safe and effective treatment, and offers beneficial outcomes. On the other hand, sub-lobar resection is as effective as lobectomy, surgery itself might have a limited role as prognostic factor. The results for the SPH analysis for central-type disease confirm that radical resection on primary tumors is an important procedure. Therefore, for central-type IIIA-SCLC patients who received NAC, pneumonectomy is associated with the best prognosis.

Analysis of the selection and use of postoperative adjuvant therapy, particularly for NAC-treated LA-SCLC patients, is the most crucial step in research to be performed next. Previously published studies have reported that standard treatments for postoperative SCLC are platinum-based chemotherapy with thoracic radiation; however, the effect of postoperative treatment remains clear (33,34). Based on the findings from the SPH patient analysis, the effect of existing postoperative adjuvant therapy is relatively limited. Adjuvant chemotherapy was not found to improve the OS of patients who received NAC plus radical resection. With the development of ICIs and AAIs for the treatment of ED-SCLC, the next step is to discuss their role in neoadjuvant and adjuvant therapy, which should be confirmed by RCTs (26,35). There is a high rate of SCLC treatment failure due to brain metastasis (13,14). Therefore, according to the current National Comprehensive Cancer Network guidelines, chemoradiotherapy in combination with adjuvant prophylactic cranial irradiation (PCI) is recommended as the standard treatment for IIIA-SCLC (7,36). The effect of adjuvant radiotherapy and PCI for NAC plus radical resection IIIA-SCLC patients warrants further research (37).

Limitations

This retrospective study based on SEER and SPH database had some limitations inevitably. First, the information of dosage and cycle of chemotherapy could not be enrolled in SEER. Second, SEER would not contain the information on whether surgery or chemotherapy was administered first. Third, we would not know the reason clearly why the proportion of patients underwent sub-lobal resection was high in SEER. Fourth, the sample size of SPH is small relatively, which limits the possibility of utilizing more advanced statistical methods and discussing more prognostic factors for LA-SCLC, especially PCI and radiotherapy. In next work, prospective clinical trials concerning NAC or chemotherapy in combination with immunotherapy followed by radical surgery for LA-SCLC has been conducted in our center. We attempted to explain the confusions caused by these limitations by the next work.


Conclusions

Compared with non-surgical regimens, NAC followed by complete resection might improve the prognosis for IIIA-SCLC patients. After NAC treatment, IIIA-SCLC, particularly primary tumors with consistent response to NAC, surgical treatment should be performed surgical treatment as long as the patients can tolerate it. The efficacy of radical sub-lobectomy seems to be inferior compared with lobectomy alone for NAC-treated IIIA-SCLC. For central-type disease, NAC followed by intentional radical primary tumor resection is a suitable therapeutic approach. Further studies on postoperative adjuvant treatments are warranted, and relevant conclusions should be verified by RCTs.


Acknowledgments

The authors appreciate the academic support from the AME Thoracic Surgery Collaborative Group.

Funding: The present study was supported by the National Natural Science Foundation of China (Grant No. 81900078).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-545/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-545/dss

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-545/coif). AA reports advisory board: MSD Oncology, Roche, Takeda, Pfizer, Bristol-Myers Squibb, AstraZeneca, Eli-Lilly; speaker bureau from Eli-Lilly, AstraZeneca, Amgen; research funding from Boehringer Ingelheim, AstraZeneca, Bristol Myers Squibb; expert testimony from Roche, AstraZeneca, Bristol Myers Squibb; travels, accommodations, expenses from Bristol Myers Squibb, AstraZeneca, Amgen; outside the submitted work. FM received research institutional grant from Astrazeneca; consulting fees and support for attending meetings and/or travel from AstraZeneca, BrainLab, Elekta; honoraria from AstraZeneca, Novartis, Roche, Lilly, BrainLab, Elekta; and participated on a Data Safety Monitoring Board or Advisory Board of AstraZeneca, Novartis; outside the submitted work. The other 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 (as revised in 2013). The study was approved by Shanghai Pulmonary Hospital ethics committee (No. K18-066), and informed consent was taken from all the patients.

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. Sabari JK, Lok BH, Laird JH, et al. Unravelling the biology of SCLC: implications for therapy. Nat Rev Clin Oncol 2017;14:549-61. [Crossref] [PubMed]
  2. 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]
  3. Che K, Shen H, Qu X, et al. Survival Outcomes for Patients with Surgical and Non-Surgical Treatments in Stages I-III Small-Cell Lung Cancer. J Cancer 2018;9:1421-9. [Crossref] [PubMed]
  4. Kalemkerian GP, Akerley W, Bogner P, et al. Small cell lung cancer. J Natl Compr Canc Netw 2013;11:78-98. [Crossref] [PubMed]
  5. Fox W, Scadding JG. Medical Research Council comparative trial of surgery and radiotherapy for primary treatment of small-celled or oat-celled carcinoma of bronchus. Ten-year follow-up. Lancet 1973;2:63-5. [Crossref] [PubMed]
  6. Govindan R, Page N, Morgensztern D, et al. Changing epidemiology of small-cell lung cancer in the United States over the last 30 years: analysis of the surveillance, epidemiologic, and end results database. J Clin Oncol 2006;24:4539-44. [Crossref] [PubMed]
  7. Ettinger DS, Wood DE, Aggarwal C, et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 1.2020. J Natl Compr Canc Netw 2019;17:1464-72. [Crossref] [PubMed]
  8. Rossi A, Di Maio M, Chiodini P, et al. Carboplatin- or cisplatin-based chemotherapy in first-line treatment of small-cell lung cancer: the COCIS meta-analysis of individual patient data. J Clin Oncol 2012;30:1692-8. [Crossref] [PubMed]
  9. Shepherd FA, Crowley J, Van Houtte P, et al. The International Association for the Study of Lung Cancer lung cancer staging project: proposals regarding the clinical staging of small cell lung cancer in the forthcoming (seventh) edition of the tumor, node, metastasis classification for lung cancer. J Thorac Oncol 2007;2:1067-77. [Crossref] [PubMed]
  10. Jin K, Zhang K, Zhou F, et al. Selection of candidates for surgery as local therapy among early-stage small cell lung cancer patients: a population-based analysis. Cancer Commun (Lond) 2018;38:5. [Crossref] [PubMed]
  11. Li S, Jin K, Pan Y, et al. Role of surgery in a case-control study of patients with clinical stage IIIA small cell lung cancer. J Thorac Dis 2021;13:2738-45. [Crossref] [PubMed]
  12. Ye P, Guo Z, Zhang Y, et al. Surgery Plus Chemotherapy Versus Surgery Alone for Limited-Stage Small-Cell Lung Cancer: A Population-Based Survival Outcome Analysis. Front Oncol 2021;11:676598. [Crossref] [PubMed]
  13. Siegel R, Ma J, Zou Z, et al. Cancer statistics, 2014. CA Cancer J Clin 2014;64:9-29. [Crossref] [PubMed]
  14. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7-30. [Crossref] [PubMed]
  15. Gelsomino F, Lamberti G, Parisi C, et al. The evolving landscape of immunotherapy in small-cell lung cancer: A focus on predictive biomarkers. Cancer Treat Rev 2019;79:101887. [Crossref] [PubMed]
  16. Lad T, Piantadosi S, Thomas P, et al. A prospective randomized trial to determine the benefit of surgical resection of residual disease following response of small cell lung cancer to combination chemotherapy. Chest 1994;106:320S-3S. [Crossref] [PubMed]
  17. Eberhardt W, Stamatis G, Stuschke M, et al. Prognostically orientated multimodality treatment including surgery for selected patients of small-cell lung cancer patients stages IB to IIIB: long-term results of a phase II trial. Br J Cancer 1999;81:1206-12. [Crossref] [PubMed]
  18. Gridelli C, Rossi A, Airoma G, et al. Treatment of pulmonary neuroendocrine tumours: state of the art and future developments. Cancer Treat Rev 2013;39:466-72. [Crossref] [PubMed]
  19. Yang CF, Chan DY, Speicher PJ, et al. Role of Adjuvant Therapy in a Population-Based Cohort of Patients With Early-Stage Small-Cell Lung Cancer. J Clin Oncol 2016;34:1057-64. [Crossref] [PubMed]
  20. Lim E, Belcher E, Yap YK, et al. The role of surgery in the treatment of limited disease small cell lung cancer: time to reevaluate. J Thorac Oncol 2008;3:1267-71. [Crossref] [PubMed]
  21. Welsh JW, Heymach JV, Guo C, et al. Phase 1/2 Trial of Pembrolizumab and Concurrent Chemoradiation Therapy for Limited-Stage SCLC. J Thorac Oncol 2020;15:1919-27. [Crossref] [PubMed]
  22. Arriola E, González-Cao M, Domine M, et al. Addition of Immune Checkpoint Inhibitors to Chemotherapy vs Chemotherapy Alone as First-Line Treatment in Extensive-Stage Small-Cell Lung Carcinoma: A Systematic Review and Meta-Analysis. Oncol Ther 2022;10:167-84. [Crossref] [PubMed]
  23. Liu Y, Cheng Y, Li K, et al. Effect of prior thoracic radiotherapy on prognosis in relapsed small cell lung cancer patients treated with anlotinib: a subgroup analysis of the ALTER 1202 trial. Transl Lung Cancer Res 2021;10:3793-806. [Crossref] [PubMed]
  24. He Z, Zhou H, Wang J, et al. Apatinib with etoposide capsules as a third- or further-line therapy for extensive-stage small cell lung cancer: an open-label, multicenter, single-arm phase II trial. Transl Lung Cancer Res 2021;10:889-99. [Crossref] [PubMed]
  25. Yu H, Chen P, Cai X, et al. Efficacy and safety of PD-L1 inhibitors versus PD-1 inhibitors in first-line treatment with chemotherapy for extensive-stage small-cell lung cancer. Cancer Immunol Immunother 2022;71:637-44. [Crossref] [PubMed]
  26. 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]
  27. 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]
  28. Yang L, Zhou Y, Wang G, et al. Clinical features and prognostic factors of combined small cell lung cancer: development and validation of a nomogram based on the SEER database. Transl Lung Cancer Res 2021;10:4250-65. [Crossref] [PubMed]
  29. Stahel R, Thatcher N, Früh M, et al. 1st ESMO Consensus Conference in lung cancer; Lugano 2010: small-cell lung cancer. Ann Oncol 2011;22:1973-80. [Crossref] [PubMed]
  30. Zhang J, Zhang L, Luo J, et al. Comprehensive genomic profiling of combined small cell lung cancer. Transl Lung Cancer Res 2021;10:636-50. [Crossref] [PubMed]
  31. Combs SE, Hancock JG, Boffa DJ, et al. Bolstering the case for lobectomy in stages I, II, and IIIA small-cell lung cancer using the National Cancer Data Base. J Thorac Oncol 2015;10:316-23. [Crossref] [PubMed]
  32. Varlotto JM, Recht A, Flickinger JC, et al. Lobectomy leads to optimal survival in early-stage small cell lung cancer: a retrospective analysis. J Thorac Cardiovasc Surg 2011;142:538-46. [Crossref] [PubMed]
  33. Grønberg BH, Killingberg KT, Fløtten Ø, et al. High-dose versus standard-dose twice-daily thoracic radiotherapy for patients with limited stage small-cell lung cancer: an open-label, randomised, phase 2 trial. Lancet Oncol 2021;22:321-31. [Crossref] [PubMed]
  34. Faivre-Finn C, Snee M, Ashcroft L, et al. Concurrent once-daily versus twice-daily chemoradiotherapy in patients with limited-stage small-cell lung cancer (CONVERT): an open-label, phase 3, randomised, superiority trial. Lancet Oncol 2017;18:1116-25. [Crossref] [PubMed]
  35. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 2017;389:255-65. [Crossref] [PubMed]
  36. Bernhardt EB, Jalal SI. Small Cell Lung Cancer. Cancer Treat Res 2016;170:301-22. [Crossref] [PubMed]
  37. Zhou N, Bott M, Park BJ, et al. Predictors of survival following surgical resection of limited-stage small cell lung cancer. J Thorac Cardiovasc Surg 2021;161:760-771.e2. [Crossref] [PubMed]
Cite this article as: Bian D, Jiang S, Xiong Y, Qi M, Wu J, Addeo A, Yamauchi Y, Manapov F, Dempke WCM, Vannucci J, Di Federico A, Xu X, Chen L. Efficacy evaluation of surgery combined with chemotherapy for stage IIIA small cell lung cancer patients: a retrospective analysis. Transl Lung Cancer Res 2022;11(8):1631-1642. doi: 10.21037/tlcr-22-545

Download Citation