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

Open Access 01.12.2023 | Research

Baseline serum tumor markers predict the survival of patients with advanced non-small cell lung cancer receiving first-line immunotherapy: a multicenter retrospective study

verfasst von: Jian Huang, Yi Xiao, Yubin Zhou, Huiyin Deng, Zihao Yuan, Longyan Dong, Jun Lan, Xiane Li, Gaijiao Liu, Hao Hu, Shaohong Huang, Xiongwen Yang

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Background

This study aimed to investigate the association between baseline serum tumor markers (STMs) (carcinoembryonic antigen [CEA], neuron-specific enolase [NSE], cytokeratin-19 fragment [CYFRA21-1], carbohydrate antigen 19–9 [CA19-9], and carbohydrate antigen 125 [CA125]) and the efficacy of first-line immunotherapy in patients with advanced non-small cell lung cancer.

Methods

This multicenter retrospective study evaluated patients who received first-line immunotherapy between July 2017 and July 2022. The endpoints were progression-free survival (PFS) and overall survival (OS), as defined by the Response Evaluation Criteria in Solid Tumors version 1.1. We divided the patients into three groups based on STM levels: Group A ≥ threefold upper limit of normal, threefold upper limit of normal > Group B > upper limit of normal, and Group C ≤ upper limit of normal.

Results

In total, 716 patients were included in this study. In Cox proportional hazards analyses, the STM levels in Group C were independently associated with superior PFS and OS in patients with lung adenocarcinoma (LUAD). Except for CA19-9 level, the STM levels in Group C were independently associated with superior PFS and OS in patients with lung squamous carcinoma (LUSC). Except for CEA and CA19-9 levels, the levels in Group A were independently associated with inferior PFS and OS in patients with LUAD and LUSC.

Conclusions

Serum CEA, NSE, CYFRA21-1, and CA125 levels can predict PFS and OS in patients with LUAD and LUSC, and serum CA19-9 levels can predict PFS and OS in patients with LUAD. The higher the serum NSE, CYFRA21-1, and CA125 levels, the worse the PFS and OS in patients with LUAD and LUSC. In addition, the higher the serum CA19-9 level, the worse the OS in patients with LUAD.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-023-11312-4.
Jian Huang, Yi Xiao and Yubin Zhou contributed equally to this work.

Publisher’s Note

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

Introduction

Lung cancer remains the main cause of death in patients with cancer, and the 5-year survival rate of patients with lung cancer is only approximately 15% [1, 2]. In the past, platinum-based chemotherapy has often been used to treat advanced lung cancer lacking the driving genes. With the advancement of immunotherapy in lung cancer, researchers have found that compared with traditional platinum-based chemotherapy strategies, immunotherapy can have better survival benefits to patients with advanced lung cancer who lack the driving genes [36]. Nevertheless, immunotherapy often fails because of tumor progression, and some patients do not benefit from immunotherapy [7]. Programmed cell death protein-1/programmed apoptosis ligand 1 (PD-L1) is the most widely known in the clinical application of immunotherapy for advanced lung cancer. The expression level of PD-L1 in patients with advanced lung cancer can be used to predict whether patients can benefit from immunotherapy [8, 9]. Nonetheless, the role of PD-L1 in predicting the prognosis of advanced lung cancer by immunotherapy is rather limited, with a single index, and the higher the expression of PD-L1, the better the prognosis of patients. Other common prediction methods include tumor mutational burden and circular tumor DNA, but these inevitably increase the extra cost of patients [10, 11].
Tumor targets are substances that exist in malignant tumor cells or are produced abnormally by malignant tumor cells. They can reflect the occurrence and development of tumors. Baseline levels of tumor markers, including carcinoembryonic antigen (CEA), cytokeratin-19 fragment (CYFRA 21–1), neuron-specific enolase (NSE), carbohydrate antigen 19–9 (CA19-9), and carbohydrate antigen 125 (CA125), have been proven to be associated with the prognosis of patients with advanced lung cancer receiving platinum-based chemotherapy [1214]. However, the role of baseline tumor marker expression levels in predicting the prognosis of patients with advanced lung cancer receiving immunotherapy remains unknown.
This study included patients with advanced non-small cell lung cancer (NSCLC) from four cancer centers in China who received first-line immunotherapy. This study primarily aimed to evaluate the predictive effect of baseline serum tumor marker (STM) levels in patients with advanced NSCLC receiving first-line immunotherapy.

Methods

Study design

We retrospectively reviewed data from 716 patients with advanced NSCLC treated with first-line immunotherapy at the Third Affiliated Hospital of Sun Yat-sen University, General Hospital of Southern Theater Command, the Third Affiliated Xiangya Hospital of Central South University, and Jiangxi Cancer Hospital between July 2017 and July 2021.

Inclusion criteria

The inclusion criteria were as follows: histologically confirmed NSCLC; diagnosis of stage IIIB to IV, including postoperative recurrence based on the eighth edition tumor-node-metastasis staging of the International Lung Cancer Research Association [15]; Eastern Cooperative Oncology Group performance status of 0–2; treatment with first-line immunotherapy; and adequate organ functions. The pathological diagnoses were performed according to the World Health Organization classification criteria [16].
Immunotherapy included immune checkpoint inhibitors with or without chemotherapy. The immune checkpoint inhibitors included pembrolizumab, nivolumab, atezolizumab, sintilimab, camrelizumab, and tirelizumab. Chemotherapy regimens included platinum-based regimens with or without bevacizumab. The duration of immunotherapy was at least 6 weeks.
Progression-free survival (PFS) was calculated from the initiation of treatment to definite tumor progression, death, or the last follow-up. Overall survival (OS) was calculated from the initiation of treatment to the date of death or last follow-up. All follow-up data were collected until October 31, 2022.
Tumor progression was assessed using the Response Evaluation Criteria in Solid Tumors version 1.1 [17], including complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD). Objective response rate (ORR) was defined as the percentage of CR + PR after immunotherapy. Disease control rate (DCR) was defined as the percentage of CR + PR + SD after immunotherapy. Efficacy was evaluated independently by two experienced physicians. Considering the possibility of pseudoprogression in immunotherapy, determination of disease progression requires two consecutive radiological examinations.
STM (CEA, NSE, CYFRA 21–1, CA19-9, and CA125) concentrations were measured at the time of diagnosis. For the reported cohort, STM analyses were performed using a cobas e 801 immunoassay module (Roche Diagnostics, Rotkreuz, Switzerland) and the corresponding ElectroChemiLuminescence-ImmunoAssay kits acquired from Roche. According to the manufacturer’s instructions, the normal upper limits of CEA, NSE, CYFRA 21–1, CA19-9, and CA125 for the diagnosis of NSCLC are 5.00, 16.3, 3.30, 27.0, and 35.0 ng/mL, respectively.
We divided the patients into three groups according to the baseline STM levels. Group A had baseline STM levels greater than three times the upper limit of the normal value. In Group B, the baseline STM levels were higher than the upper limit of the normal value and less than three times the upper limit of the normal value. In Group C, the baseline STM levels were lower than the upper limit of normal.

Statistical analyses

Continuous variables are presented as mean ± standard deviation, and categorical variables are presented as numbers (%). Categorical variables were compared using the chi-squared or Fisher’s exact test. Survival was estimated using the Kaplan–Meier method. A log-rank test was performed to evaluate the significance of the differences in survival periods among the groups. The median, 95% confidence intervals (CIs), and P values from the log-rank tests are reported in the figures. The Cox proportional hazards regression model was used for univariate and multivariate analyses to assess the prognostic role of STMs, adjusted for the possible confounding effect of all other factors included in the same model. All P values were two-sided, and values < 0.05 were considered statistically significant. Statistical analyses were performed using the R software version 4.2.1 (https://​www.​r-project.​org/​).

Results

Patient selection and characteristics

The flow diagram of the patients included in the analysis is shown in Fig. 1. Clinicopathological characteristics of the patients are shown in Table 1. In total, 716 patients with advanced NSCLC were included in this study, including 390 patients with lung adenocarcinoma (LUAD), 280 with lung squamous carcinoma (LUSC), and 46 with other types of NSCLC. The mean age of the included patients was 61.1 years, and the majority of the patients were male. In total, 605 patients had stage IV disease. Smoking and previous smoking accounted for the majority of the patients. Only a small proportion of patients were negative for PD-L1 expression. Most patients received immune checkpoint blockades in combination with chemotherapy. The mean CEA level was 57.29 ng/mL, and 409 (57.1%) patients had CEA levels above the upper limit of normal. The mean NSE level was 24.26 ng/mL, and 414 (57.8%) patients had NSE levels above the upper limit of normal. The mean CYFRA21-1 level was 15.69 ng/mL, and 618 (86.3%) patients had CYFRA21-1 levels above the upper limit of normal. The mean CA19-9 level was 45.99 ng/mL, and 268 (37.4%) patients had CA19-9 levels above the upper limit of normal. The mean CA125 level was 69.04 ng/mL, and 416 (58.1%) patients had CA125 levels above the upper limit of normal.
Table 1
Characteristics of patients at baseline
Characteristics
Patients (n = 716)
Percentage (%)
Age (Mean ± SD)
61.10 ± 10.55
 
Sex
 Male
611
85.3
 Female
105
14.8
Histological type
 LUAD
390
54.5
 LUSC
280
39.1
 Other NSCLC
46
6.4
Clinical stage
 IIIB
86
12
 IIIC
25
3.5
 IV
605
84.5
Smoking history
 Never smoker
324
45.3
 Smoker or ex-smoker
392
54.7
PD-L1 expression
  < 1%
154
21.5
 1%-49%
298
41.6
  ≥ 50%
264
36.9
Treatment type
 Monotherapy
284
39.7
 Combination therapy
432
60.3
ECOG PS
 0–1
645
90.1
 2
71
9.9
Radiation history
 Yes
452
63
 No
265
37
Metastasis sites
 Liver
59
8.24
 Lung
179
25
 Brain
135
18.9
 Bone
213
29.7
 Adrenal
124
17.3
Drug
 Pembrolizumab
451
63
 Nivolumab
108
15.1
 Atezolizumab
5
0.7
 Sintilimab
108
15.1
 Camrelizumab
31
4.3
 Tislelizumab
13
1.8
CEA (ng/ml)
 Mean ± SD
57.29 ± 159.72
 
 Normal (≤ 5.0)
307
42.9
 High (> 5.0)
409
57.1
NSE (ng/ml)
 Mean ± SD
24.26 ± 18.19
 
 Normal (≤ 16.3)
302
42.2
 High (> 16.3)
414
57.8
CYFRA21-1 (ng/ml)
 Mean ± SD
15.69 ± 27.37
 
 Normal (≤ 3.3)
98
13.7
 High (> 3.3)
618
86.3
CA19-9 (ng/ml)
 Mean ± SD
45.99 ± 106.47
 
 Normal (≤ 27.0)
448
62.6
 High (> 27.0)
268
37.4
CA125 (ng/ml)
 Mean ± SD
69.04 ± 112.97
 
 Normal (≤ 35.0)
300
41.9
 High (> 35.0)
416
58.1
LUAD Lung adenocarcinoma, LUSC Lung squamous cell carcinoma, NSCLC Non-small cell lung cancer, PD-L1 Programmed death ligand-1, CEA Carcinoembryonic antigen, NSE Neuron-specific enolase, CYFRA21-1 Cytokeratin fragment 19, CA19-9 Carbohydrate antigen 19–9, CA125 Carbohydrate antigen 125, ECOG PS Eastern cooperative oncology group performance status

Association between baseline serum tumor marker (STM) levels and survival

Analysis of the whole population

The median PFS and OS periods of the 716 patients were 398 days (95% CI, 352–540 days) and 418 days (95% CI, 678–797 days), respectively. By univariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.53 [0.44–0.66], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.40 [0.29–0.55], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.27 [0.19–0.38], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.72 [0.60–0.86], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.49 [0.36–0.66], P < 0.001), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.56 [0.41–0.77], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.59 [0.46–0.77], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.48 [0.37–0.63], P < 0.001) levels were associated with significantly different PFS among subgroups (Table 2, Fig. 2). Similarly, CEA (Group A vs. Group C, HR [95% CI] = 0.50 [0.40–0.63], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.38 [0.27–0.53], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.25 [0.17–0.35], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.66 [0.54–0.80], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.39 [0.27–0.55], P < 0.001), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.62 [0.44–0.88], P = 0.007; Group A vs. Group C, HR [95% CI] = 0.45 [0.32–0.62], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.56 [0.42–0.74], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.44 [0.33–0.59], P < 0.001) levels were associated with significantly different OS among subgroups (Table 2, Fig. 2).
Table 2
Prognostic factors for progression-free survival and overall survival in patients with advanced non-small cell lung cancer
Covariate
Univariate analysis
Multivariate analysis (CEA)
Multivariate analysis (NSE)
Multivariate analysis (CYFRA21-1)
Multivariate analysis (CA19-9)
Multivariate analysis (CA125)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
Progression-free survival
 Age
0.692
 
0.398
 
0.367
 
0.251
 
0.148
 
0.167
 
  Old (≥ 65)
 
1
 
1
 
1
 
1
 
1
 
1
  Young (< 65)
 
0.97(0.81–1.15)
 
0.92(0.77–1.11)
 
0.92(0.77–1.10)
 
0.90(0.75–1.08)
 
0.87(0.73–1.05)
 
0.88(0.73–1.06)
 Sex
0.732
 
0.33
 
0.025
 
0.226
 
0.109
 
0.137
 
  Female
 
1
 
1
 
1
 
1
 
1
 
1
  Male
 
1.05(0.81–1.34)
 
1.15(0.87–1.52)
 
1.38(1.04–1.82)
 
1.19(0.90–1.57)
 
1.25(0.95–1.65)
 
1.23(0.94–1.62)
 ECOG PS
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
  0–1
 
1
 
1
 
1
 
1
 
1
 
1
  2
 
3.06(2.36–3.97)
 
2.75(2.10–3.60)
 
2.96(2.26–3.88)
 
2.98(2.27–3.91)
 
2.94(2.24–3.86)
 
3.19(2.43–4.19)
 Smoke history
0.191
 
0.01
 
0.214
 
0.154
 
0.106
 
0.039
 
  No
 
1
 
1
 
1
 
1
 
1
 
1
  Yes
 
0.89(0.75–1.06)
 
0.77(0.63–0.94)
 
0.88(0.72–1.07)
 
0.87(0.71–1.06)
 
0.85(0.70–1.04)
 
0.81( 0.67–0.99)
 Histological type
 < 0.001
 
 < 0.001
 
 < 0.001
 
0.007
 
0.001
 
 < 0.001
 
  LUAD
 
1
 
1
 
1
 
1
 
1
 
1
  LUSC
 < 0.001
1.37(1.14–1.63)
 < 0.001
1.57(1.29–1.90)
 < 0.001
1.37(1.14–1.65)
0.018
1.26(1.04–1.53)
0.001
1.35(1.12–1.63)
 < 0.001
1.43(1.18–1.72)
  Other_NSCLC
0.003
0.50(0.32–0.79)
0.006
0.53(0.33–0.84)
 < 0.001
0.45(0.29–0.71)
0.005
0.52(0.33–0.82)
0.001
0.47(0.30–0.74)
0.004
0.51(0.32–0.80)
 Stage
0.002
 
 < 0.001
 
0.005
 
0.008
 
0.01
 
0.002
 
 IIIB
 
1
 
1
 
1
 
1
 
1
 
1
  IIIC
 < 0.001
2.67(1.63–4.37)
 < 0.001
2.52(1.51–4.23)
0.003
2.20(1.31–3.70)
0.005
2.24(1.33–3.76)
0.003
2.21(1.32–3.72)
0.001
2.32(1.38–3.91)
  IV
0.134
1.24(0.94–1.64)
0.079
1.30(0.97–1.74)
0.07
1.31(0.98–1.76)
0.042
1.09(0.82–1.47)
0.07
1.31(0.98–1.76)
0.065
1.32(0.98–1.77)
 PD-L1 expression
0.048
 
0.316
 
0.481
 
0.376
 
0.208
 
0.19
 
  < 1%
 
1
 
1
 
1
 
1
 
1
 
1
   ≥ 1%
 
0.81(0.66–1.00)
 
0.89(0.71–1.11)
 
0.92(0.74–1.15)
 
0.90(0.72–1.13)
 
0.87(0.70–1.08)
 
0.86(0.69–1.08)
 Treatment
0.133
 
0.022
 
0.038
 
0.024
 
0.133
 
0.03
 
  Monotherapy
 
1
 
1
 
1
 
1
 
1
 
1
  Combination therapy
 
0.87(0.73–1.04)
 
0.80(0.66–0.97)
 
0.82(0.67–0.99)
 
0.80(0.66–0.97)
 
0.86(0.71–1.05)
 
0.81(0.67–0.98)
 CEA
 < 0.001
 
 < 0.001
         
  3-Fold(> 15)
 
1
 
1
        
  Normal to 3-Fold (5–15)
0.163
0.86(0.69–1.07)
0.043
0.79(0.63–0.99)
        
  Normal (≤ 5.0)
 < 0.001
0.53(0.44–0.66)
 < 0.001
0.45(0.36–0.56)
        
  NSE
 < 0.001
   
 < 0.001
       
 3-Fold(> 48.9)
 
1
   
1
      
  Normal to 3-Fold (16.3–48.9)
 < 0.001
0.40(0.29–0.55)
  
 < 0.001
0.39(0.28–0.54)
      
  Normal (≤ 16.3)
 < 0.001
0.27(0.19–0.38)
  
 < 0.001
0.26(0.18–0.36)
      
  CYFRA21-1
 < 0.001
     
 < 0.001
     
 3-Fold(> 9.9)
 
1
     
1
    
  Normal to 3-Fold (3.3–9.9)
 < 0.001
0.72(0.60–0.86)
    
0.014
0.79(0.65–0.95)
    
  Normal (≤ 3.3)
 < 0.001
0.49(0.36–0.66)
    
 < 0.001
0.56(0.42–0.77)
    
  CA19-9
 < 0.001
       
 < 0.001
   
 3-Fold(> 81)
 
1
       
1
  
  Normal to 3-Fold (27–81)
0.07
0.74(0.53–1.03)
      
0.063
0.73(0.52–1.02)
  
  Normal (≤ 27.0)
 < 0.001
0.56(0.41–0.77)
      
 < 0.001
0.56(0.41–0.76)
  
 CA125
 < 0.001
         
 < 0.001
 
  3-Fold(> 105)
 
1
         
1
  Normal to 3-Fold (35–105)
 < 0.001
0.59(0.46–0.77)
        
 < 0.001
0.49(0.37–0.64)
  Normal (≤ 35.0)
 < 0.001
0.48(0.37–0.63)
        
 < 0.001
0.39(0.30–0.53)
Overall survival
 Age
0.023
 
0.013
 
0.018
 
0.005
 
0.001
 
0.002
 
  Old (≥ 65)
 
1
 
1
 
1
 
1
 
1
 
1
  Young (< 65)
 
0.80(0.66–0.97)
 
0.77(0.63–0.95)
 
0.78(0.64–0.96)
 
0.75(0.61–0.92)
 
0.72(0.59–0.89)
 
0.73(0.59–0.89)
 Sex
0.092
 
0.115
 
0.041
 
0.078
 
0.035
 
0.06
 
  Female
 
1
 
1
 
1
 
1
 
1
 
1
  Male
 
1.28(0.96–1.70)
 
1.29(0.94–1.78)
 
1.39(1.01–1.92)
 
1.33(0.97–1.83)
 
1.41(1.02–1.93)
 
1.36(0.99–1.86)
 ECOG PS
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
  0–1
 
1
 
1
 
1
 
1
 
1
 
1
  2
 
3.22(2.46–4.21)
 
3.09(2.33–4.08)
 
3.25(2.45–4.31)
 
3.31(2.50–4.40)
 
3.28(2.47–4.35)
 
3.49(2.62–4.63)
 Smoke history
0.384
 
0.484
 
0.404
 
0.984
 
0.767
 
0.806
 
  No
 
1
 
1
 
1
 
1
 
1
 
1
  Yes
 
1.09(0.90–1.32)
 
0.93(0.75–1.15)
 
1.10(0.88–1.36)
 
1.00(0.81–1.24)
 
0.97(0.78–1.20)
 
0.97(0.78–1.21)
 Histological type
 < 0.001
 
 < 0.001
 
0.005
 
0.041
 
0.001
 
 < 0.001
 
  LUAD
 
1
 
1
 
1
 
1
 
1
 
1
  LUSC
0.002
1.36(1.12–1.66)
 < 0.001
1.70(1.37–2.11)
0.001
1.40(1.14–1.72)
0.034
1.26(1.02–1.55)
0.001
1.41(1.14–1.73)
 < 0.001
1.49(1.21–1.83)
  Other_NSCLC
0.042
0.59(0.36–0.98)
0.073
0.62(0.37–1.04)
0.019
0.54(0.32–0.90)
0.085
0.64(0.38–1.06)
0.002
0.55(0.33–0.92)
0.054
0.60(0.36–1.01)
 Stage
0.034
 
0.042
 
0.234
 
0.159
 
0.162
 
0.068
 
  IIIB
 
1
 
1
 
1
 
1
 
1
 
1
  IIIC
0.006
2.10(1.23–3.57)
0.014
2.00(1.15–3.46)
0.187
1.45(0.83–2.53)
0.121
1.55(0.89–2.71)
0.075
1.66(0.95–2.90)
0.043
1.78(1.02–3.10)
  IV
0.3
1.18(0.86–1.62)
0.252
062(0.37–1.04)
0.276
1.20(0.86–1.67)
0.187
1.25(0.90–1.74)
0.265
1.21(0.87–1.68)
0.235
1.22(0.88–3.10)
 PD-L1 expression
0.001
 
0.265
 
0.209
 
0.172
 
0.087
 
0.044
 
  < 1%
 
1
 
1
 
1
 
1
 
1
 
1
   ≥ 1%
 
0.69(0.55–0.87)
 
0.87(0.69–1.11)
 
0.86(0.67–1.09)
 
0.85(0.67–1.07)
 
0.81(0.64–1.03)
 
0.78(0.62–0.99)
 Treatment
0.103
 
0.013
 
0.044
 
0.012
 
0.107
 
0.023
 
  Monotherapy
 
1
 
1
 
1
 
1
 
1
 
1
  Combination therapy
 
0.85(0.70–1.03)
 
0.76(0.62–0.94)
 
0.80(0.65–0.99)
 
0.76(0.62–0.94)
 
0.84(0.68–1.04)
 
0.78(0.63–0.97)
 CEA
 < 0.001
 
 < 0.001
         
  3-Fold(> 15)
 
1
 
1
        
  Normal to 3-Fold (5–15)
0.539
0.93(0.73–1.18)
0.139
0.83(0.65–1.06)
        
  Normal (≤ 5.0)
 < 0.001
0.50(0.40–0.63)
 < 0.001
0.42(0.32–0.54)
        
 NSE
 < 0.001
   
 < 0.001
       
  3-Fold(> 48.9)
 
1
   
1
      
  Normal to 3-Fold (16.3–48.9)
 < 0.001
0.38(0.27–0.53)
  
 < 0.001
0.40(0.29–0.56)
      
  Normal (≤ 16.3)
 < 0.001
0.25(0.17–0.35)
  
 < 0.001
0.26(0.18–0.37)
      
 CYFRA21-1
 < 0.001
     
 < 0.001
     
  3-Fold(> 9.9)
 
1
     
1
    
  Normal to 3-Fold (3.3–9.9)
 < 0.001
0.66(0.54–0.80)
    
 < 0.001
0.69(0.56–0.85)
    
  Normal (≤ 3.3)
 < 0.001
0.39(0.27–0.55)
    
 < 0.001
0.45(0.31–0.64)
    
 CA19-9
 < 0.001
       
 < 0.001
   
  3-Flod(> 81)
 
1
       
1
  
  Normal to 3-Fold (27–81)
0.007
0.62(0.44–0.88)
      
0.001
0.55(0.38–0.79)
  
  Normal (≤ 27.0)
 < 0.001
0.45(0.32–0.62)
      
 < 0.001
0.41(0.30–0.58)
  
 CA125
 < 0.001
         
 < 0.001
 
  3-Fold(> 105)
 
1
         
1
  Normal to 3-Fold (35–105)
 < 0.001
0.56(0.42–0.74)
        
 < 0.001
0.44(0.33–0.59)
  Normal (≤ 35.0)
 < 0.001
0.44(0.33–0.59)
        
 < 0.001
0.35(0.26–0.48)
ECOG PS Eastern cooperative oncology group performance status, PD-L1 Programmed death ligand-1, CEA Carcinoembryonic antigen, NSE Neuron-specific enolase, CYFRA21-1 Cytokeratin fragment 19, CA19-9 Carbohydrate antigen 19–9, CA125 Carbohydrate antigen 125
By multivariate analysis, CEA (Group A vs. Group B, HR [95% CI] = 0.79 [0.63–0.99], P = 0.043; Group A vs. Group C, HR [95% CI] = 0.45 [0.36–0.56], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.39 [0.28–0.54], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.26 [0.18–0.36], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.79 [0.65–0.95], P = 0.014; Group A vs. Group C, HR [95% CI] = 0.56 [0.42–0.77], P < 0.001), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.56 [0.41–0.76], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.49 [0.37–0.64], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.39 [0.30–0.53], P < 0.001) levels were associated with significantly different PFS among subgroups (Table 2). Similarly, by multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.42 [0.32–0.54], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.40 [0.29–0.56], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.26 [0.18–0.37], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.69 [0.56–0.85], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.45 [0.31–0.64], P < 0.001), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.55 [0.38–0.79], P = 0.001; Group A vs. Group C, HR [95% CI] = 0.41 [0.30–0.58], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.44 [0.33–0.59], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.35 [0.26–0.48], P < 0.001) levels were associated with significantly different OS among subgroups (Table 2).

Analysis of patients with lung adenocarcinoma

Overall, the median PFS and OS periods of the 390 patients with LUAD were 446 days (95% CI, 372–528 days) and 760 days (95% CI, 704–931 days), respectively. By univariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.38 [0.28–0.52], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.37 [0.24–0.58], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.24 [0.15–0.39], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.75 [0.58–0.98], P = 0.033; Group A vs. Group C, HR [95% CI] = 0.57 [0.37–0.86], P = 0.007), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.67 [0.44–0.99], P = 0.042; Group A vs. Group C, HR [95% CI] = 0.43 [0.30–0.64], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.70 [0.51–0.96], P = 0.026; Group A vs. Group C, HR [95% CI] = 0.41 [0.29–0.59], P < 0.001) levels in patients with LUAD were associated with significantly different PFS among subgroups (Table 3, Fig. 3). Similarly, CEA (Group A vs. Group C, HR [95% CI] = 0.33 [0.23–0.48], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.37 [0.24–0.59], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.22 [0.14–0.36], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.69 [0.52–0.92], P = 0.012; Group A vs. Group C, HR [95% CI] = 0.38 [0.23–0.64], P < 0.001), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.54 [0.35–0.82], P = 0.004; Group A vs. Group C, HR [95% CI] = 0.33 [0.22–0.49], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.62 [0.43–0.88], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.38 [0.25–0.56], P < 0.001) levels in patients with LUAD were associated with significantly different OS among subgroups (Table 3, Fig. 3).
Table 3
Prognostic factors for progression-free survival and overall survival in patients with lung adenocarcinoma
Covariate
Univariate analysis
Multivariate analysis (CEA)
Multivariate analysis (NSE)
Multivariate analysis (CYFRA21-1)
Multivariate analysis (CA19-9)
Multivariate analysis (CA125)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
Progression-free survival
 Age
0.458
 
0.415
 
0.251
 
0.277
 
0.057
 
0.106
 
  Old (≥ 65)
 
1
 
1
 
1
 
1
 
1
 
1
  Young (< 65)
 
0.91(0.72–1.16)
 
0.90(0.70–1.16)
 
0.86(0.67–1.11)
 
0.87(0.68–1.12)
 
0.78(0.61–1.01)
 
0.81(0.63–1.05)
 Sex
0.238
 
0.736
 
0.463
 
0.655
 
0.967
 
0.808
 
  Female
 
1
 
1
 
1
 
1
 
1
 
1
  Male
 
0.81(0.56–1.15)
 
1.08(0.72–1.60)
 
1.16(0.78–1.75)
 
1.10(0.73–1.64)
 
0.99(0.67–1.47)
 
105(0.71–1.56)
 ECOG PS
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
  0–1
 
1
 
1
 
1
 
1
 
1
 
1
  2
 
3.86(2.71–5.50)
 
3.34(2.31–4.84)
 
3.41(2.34–4.97)
 
3.96(2.72–5.76)
 
3.77(2.60–5.45)
 
4.29(2.94–6.26)
 Smoke history
 < 0.001
 
 < 0.001
 
0.016
 
0.005
 
0.008
 
 < 0.001
 
  No
 
1
 
1
 
1
 
1
 
1
 
1
  Yes
 
0.65(0.51–0.83)
 
0.62(0.48–0.81)
 
0.72(0.55–0.94)
 
0.69(0.53–0.90)
 
0.70(0.54–0.91)
 
0.62(0.48–0.81)
 Stage
0.193
 
0.043
 
0.512
 
0.482
 
0.565
 
0.351
 
  IIIB
 
1
 
1
 
1
 
1
 
1
 
1
  IIIC
0.167
1.77(0.79–3.99)
0.029
2.77(1.11–6.89)
0.708
1.18(0.49–2.85)
0.562
1.30(0.53–3.18)
0.565
1.29(0.54–3.10)
0.368
1.50(0.62–3.62)
  IV
0.573
0.88(0.57–1.37)
0.868
0.96(0.61–1.52)
0.358
0.75(0.58–0.97)
0.432
0.83(0.53–1.31)
0.4
0.82(0.52–1.30)
0.333
0.80(0.51–1.26)
 PD-L1 expression
0.032
 
0.413
 
0.448
 
0.246
 
0.439
 
0.624
 
  < 1%
 
1
 
1
 
1
 
1
 
1
 
1
   ≥ 1%
 
0.72(0.53–0.97)
 
0.88(0.64–1.22)
 
0.88(0.63–1.22)
 
0.83(0.60–1.14)
 
0.88(0.64–1.21)
 
0.92(0.67–1.27)
 Treatment
0.02
 
0.002
 
0.032
 
0.006
 
0.044
 
0.017
 
  Monotherapy
 
1
 
1
 
1
 
1
 
1
 
1
  Combination therapy
 
0.75(0.59–0.95)
 
0.67(0.5–0.87)
 
0.75(0.58–0.97)
 
0.69(0.53–0.90)
 
0.76(0.59–0.99)
 
0.72(0.55–0.94)
 CEA
 < 0.001
 
 < 0.001
         
  3-Fold(> 15)
 
1
 
1
        
  Normal to 3-Fold (5–15)
0.246
0.85(0.64–1.12)
0.137
0.81(0.61–1.07)
        
  Normal (≤ 5.0)
 < 0.001
0.38(0.28–0.52)
 < 0.001
0.33(0.23–0.46)
        
 NSE
 < 0.001
   
 < 0.001
       
 3-Fold(> 48.9)
 
1
   
1
      
  Normal to 3-Fold (16.3–48.9)
 < 0.001
0.37(0.24–0.58)
  
 < 0.001
0.41(0.26–0.66)
      
  Normal (≤ 16.3)
 < 0.001
0.24(0.15–0.39)
  
 < 0.001
0.29(0.18–0.47)
      
 CYFRA21-1
0.011
     
0.004
     
  3-Fold(> 9.9)
 
1
     
1
    
  Normal to 3-Fold (3.3–9.9)
0.033
0.75(0.58–0.98)
    
0.006
0.69(0.53–0.90)
    
  Normal (≤ 3.3)
0.007
0.57(0.37–0.86)
    
0.003
0.53(0.35–0.80)
    
 CA19-9
 < 0.001
       
 < 0.001
   
  3-Fold(> 81)
 
1
       
1
  
  Normal to 3-Fold (27–81)
0.042
0.67(0.44–0.99)
      
0.063
0.68(0.45–1.02)
  
  Normal (≤ 27.0)
 < 0.001
0.43(0.30–0.64)
      
 < 0.001
0.45(0.31–0.67)
  
 CA125
 < 0.001
         
 < 0.001
 
  3-Fold(> 105)
 
1
         
1
  Normal to 3-Fold (35–105)
0.026
0.70(0.51–0.96)
        
 < 0.001
0.54(0.39–0.76)
  Normal (≤ 35.0)
 < 0.001
0.41(0.29–0.59)
        
 < 0.001
0.31(0.21–0.44)
Overall survival
 Age
0.132
 
0.057
 
0.102
 
0.085
 
0.008
 
0.017
 
  Old (≥ 65)
 
1
 
1
 
1
 
1
 
1
 
1
  Young (< 65)
 
0.81(0.62–1.07)
 
0.76(0.57–1.01)
 
0.79(0.59–1.05)
 
0.78(0.59–1.03)
 
0.68(0.51–0.90)
 
0.71(0.53–0.94)
 Sex
0.673
 
0.728
 
0.527
 
0.351
 
0.87
 
0.675
 
  Female
 
1
 
1
 
1
 
1
 
1
 
1
  Male
 
0.92(0.62–1.36)
 
1.08(0.69–1.69)
 
1.15(0.74–1.82)
 
1.24(0.79–1.94)
 
1.04(0.67–1.61)
 
1.11(0.71–1.72)
 ECOG PS
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
  0–1
 
1
 
1
 
1
 
1
 
1
 
1
  2
 
4.11(2.87–5.87)
 
3.61(2.45–5.33)
 
3.69(2.49–5.46)
 
4.43(2.99–6.56)
 
4.31(2.93–6.35)
 
4.76(3.20–7.08)
Smoke history
0.089
 
0.117
 
0.653
 
0.166
 
0.219
 
0.102
 
  No
 
1
 
1
 
1
 
1
 
1
 
1
  Yes
 
0.79(0.60–1.04)
 
0.79(0.59–1.06)
 
1.45(0.55–3.80)
 
0.81(0.61–1.09)
 
0.83(0.62–1.12)
 
0.78(0.58–1.05)
 Stage
0.169
 
0.034
 
0.45
 
0.674
 
0.657
 
0.275
 
  IIIB
 
1
 
1
 
1
 
1
 
1
 
1
  IIIC
0.059
2.34(0.97–5.65)
0.007
4.01(1.17–10.94)
0.884
0.93(0.69–1.26)
0.261
1.75(0.66–4.64)
0.321
1.62(0.62–4.22)
0.902
1.71(0.65–4.48)
  IV
0.682
1.13(0.64–1.98)
0.519
1.21(0.68–2.17)
0.922
1.03(0.57–1.85)
0.832
1.07(0.60–1.91)
0.869
1.06(0.59–1.88)
0.831
0.94(0.53–1.68)
 PD-L1 expression
0.037
 
0.756
 
0.867
 
0.831
 
0.759
 
0.935
 
  < 1%
 
1
 
1
 
1
 
1
 
1
 
1
  ≥ 1%
 
0.71(0.51–0.98)
 
1.06(0.74–1.51)
 
1.10(0.77–1.58)
 
0.96(0.68–1.36)
 
1.05(0.74–1.51)
 
1.02(0.71–1.45)
Treatment
0.122
 
0.001
 
0.019
 
0.003
 
0.036
 
0.009
 
  Monotherapy
 
1
 
1
 
1
 
1
 
1
 
1
  Combination therapy
 
0.81(0.61–1.06)
 
0.62(0.46–0.82)
 
0.70(0.52–0.94)
 
0.65(0.48–0.77)
 
0.73(0.55–0.98)
 
0.67(0.50–0.91)
 CEA
 < 0.001
 
 < 0.001
         
  3-Fold(> 15)
 
1
 
1
        
  Normal to 3-Fold (5–15)
0.523
0.90(0.66–1.23)
0.309
0.85(0.62–1.16)
        
  Normal (≤ 5.0)
 < 0.001
0.33(0.23–0.48)
 < 0.001
0.28(0.18–0.42)
        
 NSE
 < 0.001
   
 < 0.001
       
  3-Fold(> 48.9)
 
1
   
1
      
  Normal to 3-Fold (16.3–48.9)
 < 0.001
0.37(0.24–0.59)
  
 < 0.001
0.42(0.26–0.68)
      
  Normal (≤ 16.3)
 < 0.001
0.22(0.14–0.36)
  
 < 0.001
0.27(0.16–0.46)
      
 CYFRA21-1
 < 0.001
     
 < 0.001
     
  3-Fold(> 9.9)
 
1
     
1
    
  Normal to 3-Fold (3.3–9.9)
0.012
0.69(0.52–0.92)
    
 < 0.001
0.58(0.43–0.77)
    
  Normal (≤ 3.3)
 < 0.001
0.38(0.23–0.64)
    
 < 0.001
0.37(0.22–0.62)
    
 CA19-9
 < 0.001
       
 < 0.001
   
  3-Fold(> 81)
 
1
       
1
  
  Normal to 3-Fold (27–81)
0.004
0.54(0.35–0.82)
      
 < 0.001
0.48(0.31–0.73)
  
  Normal (≤ 27.0)
 < 0.001
0.33(0.22–0.49)
      
 < 0.001
0.29(0.19–0.44)
  
 CA125
 < 0.001
         
 < 0.001
 
  3-Fold(> 105)
 
1
         
1
  Normal to 3-Fold (35–105)
0.007
0.62(0.43–0.88)
        
 < 0.001
0.47(0.33–0.68)
  Normal (≤ 35.0)
 < 0.001
0.38(0.25–0.56)
        
 < 0.001
0.28(0.19–0.42)
ECOG PS Eastern cooperative oncology group performance status, PD-L1 Programmed death ligand-1, CEA Carcinoembryonic antigen, NSE Neuron-specific enolase, CYFRA21-1 Cytokeratin fragment 19, CA19-9 Carbohydrate antigen 19–9, CA125 Carbohydrate antigen 125
By multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.33 [0.23–0.46], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.41 [0.26–0.66], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.29 [0.18–0.47], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.69 [0.53–0.90], P = 0.006; Group A vs. Group C, HR [95% CI] = 0.53 [0.35–0.80], P = 0.003), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.45 [0.31–0.67], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.54 [0.39–0.76], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.31 [0.21–0.44], P < 0.001) levels in patients with LUAD were associated with significantly different PFS among subgroups (Table 3). Similarly, CEA (Group A vs. Group C, HR [95% CI] = 0.28 [0.18–0.42], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.42 [0.26–0.68], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.27 [0.16–0.46], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.58 [0.43–0.77], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.37 [0.22–0.62], P < 0.001), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.48 [0.31–0.73], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.29 [0.19–0.44], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.47 [0.33–0.68], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.28 [0.19–0.42], P < 0.001) levels in patients with LUAD were associated with significantly different OS among subgroups (Table 3).

Analysis of patients with lung squamous carcinoma

Overall, the median PFS and OS periods of the 280 patients with LUSC were 336 days (95% CI, 292–385 days) and 623 days (95% CI, 505–759 days), respectively. By univariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.49 [0.35–0.70], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.74 [0.56–0.98], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.21 [0.13–0.36], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.74 [0.56–0.98], P = 0.035; Group A vs. Group C, HR [95% CI] = 0.44 [0.26–0.73], P = 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.44 [0.27–0.71], P = 0.01; Group A vs. Group C, HR [95% CI] = 0.45 [0.28–0.72], P < 0.001) levels in patients with LUSC were associated with significantly different PFS among subgroups (Table 4, Fig. 4). Similarly, CEA (Group A vs. Group C, HR [95% CI] = 0.39 [0.26–0.57], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.38 [0.27–0.53], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.23 [0.14–0.39], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.59 [0.44–0.81], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.38 [0.22–0.66], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.45 [0.27–0.75], P = 0.002; Group A vs. Group C, HR [95% CI] = 0.37 [0.22–0.62], P < 0.001) levels in patients with LUSC were associated with significantly different OS among subgroups (Table 4, Fig. 4).
Table 4
Prognostic factors for progression-free survival and overall survival in patients with lung squamous cell carcinoma
Covariate
Univariate analysis
Multivariate analysis (CEA)
Multivariate analysis (NSE)
Multivariate analysis (CYFRA21-1)
Multivariate analysis (CA19-9)
Multivariate analysis (CA125)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
P
HR (95% CI)
Progression-free survival
 Age
0.625
 
0.8
 
0.336
 
0.782
 
0.642
 
0.682
 
  Old (≥ 65)
 
1
 
1
 
1
 
1
 
1
 
1
  Young (< 65)
 
0.93(0.72–1.22)
 
1.04(0.77–1.39)
 
1.16(0.86–1.57)
 
1.04(0.78–1.40)
 
1.07(0.80–1.44)
 
1.06(0.79–1.43)
 Sex
0.02
 
0.186
 
0.041
 
0.314
 
0.102
 
0.158
 
  Female
 
1
 
1
 
1
 
1
 
1
 
1
  Male
 
1.52(1.06–2.17)
 
1.32(0.87–2.01)
 
1.55(1.02–2.35)
 
1.24(0.81–1.89)
 
1.41(0.93–2.14)
 
1.35(0.89–2.04)
 ECOG PS
 < 0.001
 
0.005
 
 < 0.001
 
0.004
 
0.003
 
0.001
 
  0–1
 
1
 
1
 
1
 
1
 
1
 
1
  2
 
2.30(1.52–3.48)
 
0.90(1.22–2.96)
 
2.19(1.40–3.43)
 
1.92(1.22–2.99)
 
1.96(1.26–3.07)
 
2.05(1.32–3.19)
 Smoke history
0.006
 
0.372
 
0.119
 
0.108
 
0.147
 
0.179
 
  No
 
1
 
1
 
1
 
1
 
1
 
1
  Yes
 
1.45(1.11–1.88)
 
1.17(0.83–1.63)
 
1.30(0.94–1.80)
 
1.31(0.94–1.81)
 
1.28(0.92–1.78)
 
1.25(0.90–1.77)
 Stage
 < 0.001
 
0.007
 
0.008
 
0.006
 
0.002
 
 < 0.001
 
  IIIB
 
1
   
1
   
1
 
1
  IIIC
 < 0.001
3.20(1.71–5.99)
0.002
2.82(1.46–5.47)
0.001
3.00(1.55–5.82)
0.005
2.61(1.33–5.10)
0.002
2.87(1.48–5.58)
 < 0.001
3.29(1.69–6.38)
  IV
0.005
1.70(1.17–2.45)
0.017
1.59(1.08–2.34)
0.011
1.64(1.12–2.40)
0.006
1.70(1.16–2.48)
0.006
1.70(1.16–2.50)
0.005
1.72(1.18–2.52)
 PD-L1 expression
0.774
 
0.658
 
0.485
 
0.383
 
0.665
 
0.846
 
  < 1%
 
1
 
1
 
1
 
1
 
1
 
1
  ≥ 1%
 
0.97(1.05–1.30)
 
1.08(0.77–1.51)
 
1.13(0.80–1.59)
 
1.16(0.83–1.64)
 
1.08(0.77–1.51)
 
0.97(0.69–1.36)
 Treatment
0.553
 
0.628
 
363
 
0.489
 
0.708
 
0.562
 
  Monotherapy
 
1
 
1
 
1
 
1
 
1
 
1
  Combination therapy
 
1.09(0.82–1.44)
 
0.93(0.68–1.27)
 
0.87(0.63–1.18)
 
0.90(0.65–1.23)
 
0.94(0.69–1.29)
 
0.91(0.67–1.24)
 CEA
 < 0.001
 
0.041
         
  3-Fold(> 15)
 
1
 
1
        
  Normal to 3-Fold (5–15)
0.058
0.68(0.46–1.01)
0.233
0.78(0.51–1.18)
        
  Normal (≤ 5.0)
 < 0.001
0.49(0.35–0.70)
0.023
0.64(0.44–0.94)
        
 NSE
 < 0.001
   
 < 0.001
       
  3-Fold(> 48.9)
 
1
   
1
      
  Normal to 3-Fold (16.3–48.9)
 < 0.001
0.36(0.22–0.58)
  
 < 0.001
0.31(0.19–0.52)
      
  Normal (≤ 16.3)
 < 0.001
0.21(0.13–0.36)
  
 < 0.001
0.18(0.11–0.31)
      
 CYFRA21-1
0.013
     
0.036
     
  3-Fold(> 9.9)
 
1
     
1
    
  Normal to 3-Fold (3.3–9.9)
0.035
0.74(0.56–0.98)
    
0.303
0.86(0.64–1.15)
    
  Normal (≤ 3.3)
0.001
0.44(0.26–0.73)
    
0.013
0.51(0.30–0.87)
    
 CA19-9
0.754
       
0.857
   
  3-Fold(> 81)
 
1
       
1
  
  Normal to 3-Fold (27–81)
0.911
1.03(0.57–1.89)
      
0.715
1.12(0.61–2.07)
  
  Normal (≤ 27.0)
0.553
0.84(0.48–1.49)
      
0.952
1.02(0.57–1.83)
  
 CA125
 < 0.001
         
 < 0.001
 
  3-Fold(> 105)
 
1
         
1
  Normal to 3-Fold (35–105)
 < 0.001
0.44(0.27–0.71)
        
 < 0.001
0.41(0.25–0.69)
  Normal (≤ 35.0)
 < 0.001
0.45(0.28–0.72)
        
0.004
0.48(0.29–0.79)
Overall survival
 Age
0.006
 
0.057
 
0.107
 
0.054
 
0.055
 
0.062
 
  Old (≥ 65)
 
1
 
1
 
1
 
1
 
1
 
1
  Young (< 65)
 
0.67(0.50–0.89)
 
0.73(0.53–1.01)
 
0.76(0.55–1.06)
 
0.73(0.52–1.01)
 
0.73(0.53–1.01)
 
0.73(0.53–1.02)
 Sex
0.003
 
0.068
 
0.041
 
0.147
 
0.033
 
0.053
 
  Female
 
1
 
1
 
1
 
1
 
1
 
1
  Male
 
1.88(1.24–2.85)
 
1.56(0.97–2.52)
 
1.65(1.02–2.66)
 
1.43(0.88–2.32)
 
1.56(0.95–2.56)
 
1.60(0.99–2.58)
 ECOG PS
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
 < 0.001
 
  0–1
 
1
 
1
 
1
 
1
 
1
 
1
  2
 
2.30(1.50–3.55)
 
2.36(1.49–3.73)
 
2.71(1.70–4.34)
 
2.24(1.40–3.60)
 
2.34(1.46–3.73)
 
2.46(1.55–3.90)
Smoke history
 < 0.001
 
0.542
 
0.126
 
0.104
 
0.271
 
0.244
 
  No
 
1
 
1
 
1
 
1
 
1
 
1
  Yes
 
1.77(1.33–2.36)
 
1.12(0.78–1.60)
 
1.33(0.92–1.91)
 
1.34(0.94–1.92)
 
1.22(0.85–1.76)
 
1.23(0.86–1.77)
 Stage
0.062
 
0.215
 
0.313
 
0.268
 
0.172
 
0.115
 
  IIIB
 
1
   
1
   
1
 
1
  IIIC
0.078
1.83(0.93–3.57)
0.136
1.70(0.85–3.39)
0.338
1.42(0.70–2.88)
0.333
1.42(0.69–2.90)
0.151
1.68(0.83–3.40)
0.086
1.85(0.92–3.74)
  IV
0.14
1.35(0.91–1.99)
0.291
1.24(0.83–1.86)
0.228
1.28(0.86–1.92)
0.217
1.29(0.86–1.93)
0.191
1.31(0.87–1.96)
0.173
1.32(0.88–1.97)
 PD-L1 expression
0.023
 
0.344
 
0.193
 
0.558
 
0.196
 
0.079
 
  < 1%
 
1
 
1
 
1
 
1
 
1
 
1
  ≥ 1%
 
0.68(0.49–0.95)
 
0.84(0.59–1.20)
 
0.79(0.55–1.13)
 
0.97(0.70–1.35)
 
0.79(0.55–1.13)
 
0.72(0.50–1.04)
 Treatment
0.948
 
0.847
 
0.878
 
0.879
 
0.67
 
0.768
 
  Monotherapy
 
1
 
1
 
1
 
1
 
1
 
1
  Combination therapy
 
0.99(0.73–1.35)
 
1.03(0.74–1.43)
 
1.03(0.74–1.43)
 
0.97(0.70–1.35)
 
1.07(0.77–1.50)
 
1.05(0.76–1.46)
  CEA
 < 0.001
 
 < 0.001
         
 3-Fold(> 15)
 
1
 
1
        
  Normal to 3-Fold (5–15)
0.106
0.71(0.46–1.08)
0.146
0.73(0.47–1.12)
        
  Normal (≤ 5.0)
 < 0.001
0.39(0.26–0.57)
 < 0.001
0.48(0.32–0.73)
        
  NSE
 < 0.001
   
 < 0.001
       
 3-Fold(> 48.9)
 
1
   
1
      
  Normal to 3-Fold (16.3–48.9)
 < 0.001
0.35(0.21–0.57)
  
 < 0.001
0.33(0.20–0.56)
      
  Normal (≤ 16.3)
 < 0.001
0.23(0.14–0.39)
  
 < 0.001
0.22(0.13–0.37)
      
 CYFRA21-1
 < 0.001
     
0.015
     
  3-Fold(> 9.9)
 
1
     
1
    
  Normal to 3-Fold (3.3–9.9)
 < 0.001
0.59(0.44–0.81)
    
0.028
0.70(0.51–0.96)
    
  Normal (≤ 3.3)
 < 0.001
0.38(0.22–0.66)
    
0.005
0.44(0.24–0.78)
    
 CA19-9
0.616
       
0.473
   
  3-Fold(> 81)
 
1
       
1
  
  Normal to 3-Fold (27–81)
0.84
0.94(0.49–1.80)
      
0.688
0.87(0.44–1.71)
  
  Normal (≤ 27.0)
0.238
0.69(0.37–1.28)
      
0.353
0.74(0.39–1.40)
  
 CA125
0.002
         
 < 0.001
 
  3-Fold(> 105)
 
1
         
1
  Normal to 3-Fold (35–105)
0.002
0.45(0.27–0.75)
        
 < 0.001
0.38(0.22–0.64)
  Normal (≤ 35.0)
 < 0.001
0.37(0.22–0.62)
        
 < 0.001
0.36(0.22–0.61)
ECOG PS Eastern cooperative oncology group performance status, PD-L1 Programmed death ligand-1, CEA Carcinoembryonic antigen, NSE Neuron-specific enolase, CYFRA21-1 Cytokeratin fragment 19, CA19-9 Carbohydrate antigen 19–9, CA125 Carbohydrate antigen 125
By multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.64 [0.44–0.94], P = 0.023), NSE (Group A vs. Group B, HR [95% CI] = 0.31 [0.19–0.52], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.18 [0.11–0.31], P < 0.001), CYFRA21-1 (Group A vs. Group C, HR [95% CI] = 0.51 [0.30–0.87], P = 0.013), and CA125 (Group A vs. Group B, HR [95% CI] = 0.41 [0.25–0.69], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.48 [0.29–0.79], P = 0.004) levels in patients with LUSC were associated with significantly different PFS among subgroups (Table 4). Similarly, CEA (Group A vs. Group C, HR [95% CI] = 0.48 [0.32–0.73], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.33 [0.20–0.56], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.22 [0.13–0.37], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.70 [0.51–0.96], P = 0.028; Group A vs. Group C, HR [95% CI] = 0.44 [0.24–0.78], P = 0.005), and CA125 (Group A vs. Group B, HR [95% CI] = 0.38 [0.22–0.64], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.36 [0.22–0.61], P < 0.001) levels in patients with LUSC were associated with significantly different OS among subgroups (Table 4).

Analysis of patients with non-small cell lung cancer who received monotherapy

By univariate analysis, NSE (Group A vs. Group B, HR [95% CI] = 0.44 [0.26–0.74], P = 0.002; Group A vs. Group C, HR [95% CI] = 0.16 [0.09–0.29], P < 0.001), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.44 [0.32–0.97], P = 0.038), and CA125 (Group A vs. Group C, HR [95% CI] = 0.65[0.42–0.99], P = 0.045) levels were associated with significantly different PFS among subgroups (Table S3). Similarly, NSE (Group A vs. Group B, HR [95% CI] = 0.38 [0.22–0.63], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.25 [0.15–0.43], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.71 [0.51–0.98], P = 0.039; Group A vs. Group C, HR [95% CI] = 0.49 [0.24–0.76], P = 0.004), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.56 [0.25–0.77], P = 0.004), and CA125 (Group A vs. Group C, HR [95% CI] = 0.61 [0.38–0.98], P = 0.039) levels were associated with significantly different OS among subgroups (Table S3).
By multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.60[0.42–0.86], P = 0.003), NSE (Group A vs. Group B, HR [95% CI] = 0.39[0.23–0.67], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.23[0.13–0.40], P < 0.001), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.48[0.27–0.86], P = 0.013), and CA125 (Group A vs. Group B, HR [95% CI] = 0.49 [0.37–0.64], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.43 [0.27–0.67], P < 0.001) levels were associated with significantly different PFS among subgroups (Table S3). Similarly, by multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.64[0.43–0.95], P = 0.026), NSE (Group A vs. Group B, HR [95% CI] = 0.31 [0.18–0.53], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.14[0.08–0.25), P < 0.001), CYFRA21-1 ( Group A vs. Group C, HR [95% CI] = 0.49 [0.28–0.89], P = 0.018), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.33[0.18–0.60], P < 0.001), and CA125 ( Group A vs. Group C, HR [95% CI] = 0.41 [0.25–0.68], P < 0.001) levels were associated with significantly different OS among subgroups (Table S3).

Analysis of patients with non-small cell lung cancer who received combination therapy

By univariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.40 [0.30–0.52], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.37 [0.24–0.56], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.27 [0.17–0.41], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.65 [0.52–0.82], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.40 [0.27–0.59], P < 0.001), CA19-9 (Group A vs. Group C, HR [95% CI] = 0.56[0.39–0.82], P = 0.003), and CA125 (Group A vs. Group B, HR [95% CI] = 0.38 [0.27–0.54], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.32 [0.23–0.46], P < 0.001) levels were associated with significantly different PFS among subgroups (Table S4). Similarly, NSE (Group A vs. Group B, HR [95% CI] = 0.38 [0.22–0.63], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.25 [0.15–0.43], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.61[0.47–0.79], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.35 [0.22–0.55], P < 0.001), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.59 [0.38–0.91], P = 0.018; Group A vs. Group C, HR [95% CI] = 0.46 [0.31–0.69], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.33 [0.23–0.48], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.29 [0.20–0.43], P < 0.001) levels were associated with significantly different OS among subgroups (Table S4).
By multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.36(0.27–0.48), P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.35 [0.23–0.54], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.25 [0.16–0.39], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.69 [0.54–0.88], P = 0.002; Group A vs. Group C, HR [95% CI] = 0.46 [0.31–0.68], P < 0.001), CA19-9 ( Group A vs. Group C, HR [95% CI] = 0.58 [0.39–0.85], P = 0.005), and CA125 (Group A vs. Group B, HR [95% CI] = 0.36 [0.25–0.51], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.32 [0.22–0.46], P < 0.001) levels were associated with significantly different PFS among subgroups (Table S4). Similarly, by multivariate analysis, CEA (Group A vs. Group C, HR [95% CI] = 0.32 [0.23–0.44], P < 0.001), NSE (Group A vs. Group B, HR [95% CI] = 0.47 [0.30–0.74], P = 0.001; Group A vs. Group C, HR [95% CI] = 0.36[0.23–0.58], P < 0.001), CYFRA21-1 (Group A vs. Group B, HR [95% CI] = 0.66[0.51–0.86], P = 0.002; Group A vs. Group C, HR [95% CI] = 0.41[0.26–0.66], P < 0.001), CA19-9 (Group A vs. Group B, HR [95% CI] = 0.58 [0.39–0.85], P = 0.005; Group A vs. Group C, HR [95% CI] = 0.45 [0.30–0.68], P < 0.001), and CA125 (Group A vs. Group B, HR [95% CI] = 0.63 [0.20–0.44], P < 0.001; Group A vs. Group C, HR [95% CI] = 0.28 [0.19–0.42), P < 0.001) levels were associated with significantly different OS among subgroups (Table S4).

Association between STM levels and programmed apoptosis ligand 1 expression

All patients underwent PD-L1 testing, and 154 of them were negative for PD-L1 expression (Table 1). In patients with LUAD, only CA19-9 concentration and PD-L1 expression were statistically different (Table 5). In patients with LUSC, there was no statistical difference between STM concentrations and PD-L1 expression (Table 5).
Table 5
The association with serum tumor markers and PD-L1 expression levels in patients
Serum tumor markers
LUAD (N = 390)
LUSC (N = 280)
PD-L1( +)
PD-L1 (-)
P value
PD-L1( +)
PD-L1 (-)
P value
CEA (ng/ml)
  
0.161
  
0.242
 Normal (≤ 5.0)
100
17
 
78
30
 
 High (> 5.0)
215
58
 
136
36
 
NSE (ng/ml)
  
0.675
  
1
 Normal (≤ 16.3)
137
30
 
89
27
 
 High (> 16.3)
178
45
 
125
39
 
CYFRA21-1 (ng/ml)
  
0.795
  
0.104
 Normal (≤ 3.3)
44
9
 
27
3
 
 High (> 3.3)
271
66
 
187
63
 
CA19-9 (ng/ml)
  
0.043
  
0.956
 Normal (≤ 27.0)
194
36
 
143
45
 
 High (> 27.0)
121
39
 
71
21
 
CA125 (ng/ml)
 Normal (≤ 35.0)
114
23
0.444
109
30
0.524
 High (> 35.0)
201
52
 
105
36
 
LUAD Lung adenocarcinoma, LUSC Lung squamous cell carcinoma, PD-L1 Programmed death ligand-1, CEA Carcinoembryonic antigen, NSE Neuron-specific enolase, CYFRA21-1 Cytokeratin fragment 19, CA19-9 Carbohydrate antigen 19–9, CA125 Carbohydrate antigen 125

Correlation between STM levels and tumor response

In patients with LUAD, the ORRs of the STM (CEA, NSE, CA19-9, and CA125) groups were statistically different (P < 0.001, P < 0.001, P = 0.014, and P = 0.002, respectively) (Table 6). In addition, the DCRs of the STMs = (CEA, NSE, CYFRA 21–1, and CA19-9) groups were statistically different (P < 0.001, P < 0.001, P = 0.007, and P = 0.001, respectively) (Table 6).
Table 6
The association with serum tumor markers and ORRs and DCRs
Serum tumor markers
LUAD (N = 390)
LUSC (N = 280)
CEA group
ORR
Non-ORR
P value
DCR
Non-DCR
P value
ORR
Non-ORR
P value
DCR
Non-DCR
P value
A*
52
119
 < 0.001
141
30
 < 0.001
14
33
0.269
39
8
0.006
B
51
51
 
95
7
 
23
44
 
56
11
C
70
47
 
112
5
 
69
97
 
157
9
NSE group
 A
2
22
 < 0.001
11
13
 < 0.001
7
12
0.873
14
5
0.005
 B
88
111
 
177
22
 
53
92
 
127
18
 C
83
84
 
160
7
 
46
70
 
111
5
CYFRA21-1 group
 A
50
80
0.212
108
22
0.007
49
87
0.553
117
19
0.107
 B
96
111
 
188
19
 
43
71
 
107
7
 C
27
26
 
52
1
 
14
16
 
28
2
CA19-9 group
 A
13
26
0.014
31
8
0.001
7
10
0.206
17
0
0.013
 B
44
77
 
101
20
 
22
53
 
61
14
 C
116
114
 
216
14
 
77
111
 
174
14
CA125 group
 A
20
43
0.002
53
10
0.102
4
18
0.016
16
6
0.025
 B
77
113
 
167
23
 
39
80
 
110
9
 C
76
61
 
128
9
 
63
76
 
126
13
LUAD Lung adenocarcinoma, LUSC Lung squamous cell carcinoma, CEA Carcinoembryonic antigen, NSE Neuron-specific enolase, CYFRA21-1 Cytokeratin fragment 19, CA19-9 Carbohydrate antigen 19–9, CA125 Carbohydrate antigen 125, ORR Objective response rate, DCR Disease control rate
*Group A: Level of baseline serum tumor markers greater than 3 times the upper limit of normal value
Group B: Level of baseline serum tumor markers was higher than the upper limit of normal value and lower than 3 times the upper limit of normal value
Group C: Level of baseline serum tumor markers were lower than the upper limit of normal
In patients with LUSC, the ORRs of the STM (CA125) groups were statistically different (P = 0.016), and the DCRs of the STM (CEA, NSE, CA19-9, and CA125) groups were statistically different (P = 0.006, P = 0.005, P = 0.013, and P = 0.025, respectively) (Table 6).

Discussion

Several large-scale clinical studies have confirmed the efficacy of first-line immunotherapy in improving survival in patients with advanced NSCLC [36], but screening the potential patients who can benefit from immunotherapy before treatment remains unclear [10]. PD-L1, the most commonly used biomarker to predict the efficacy of immunotherapy, has limitations [8, 9, 18].
As routinely measured clinical biomarkers, STMs have been used on a large scale in the diagnosis of malignancies and in the prediction of efficacy [1923]. To date, several studies have reported an association between baseline or dynamic STMs and the efficacy of immunotherapy [2427]. However, all these studies have limitations. First, previous studies did not specifically focus on patients receiving first-line immunotherapy and included a small number of patients, which may have led to potential bias [2427]. To the best of our knowledge, to date, our study is the largest study to investigate the association between baseline STM levels and first-line immunotherapy efficacy. Second, previous studies simply dichotomized the concentration of STMs, making it difficult to distinguish patients who can benefit from immunotherapy more precisely [2426]. In our study, we used the upper limit of the normal value of STMs and threefold the upper limit of the normal value of STMs as cut-off values to divide the STMs into three groups. This classification was used to confirm whether STM concentrations were associated with immunotherapy efficacy.
CEA, the classical and most widely used STM [28], had superior PFS and OS at baseline CEA levels, lower than the upper limit of normal in patients with LUAD and LUSC in our cohort. However, higher baseline CEA levels are not associated with worse prognosis. Therefore, in clinical practice, we do not predict patient survival based on serum CEA concentrations if they are above the upper limit of the normal value.
NSE is commonly used in small cell lung cancers [29]. Although serum NSE level is also elevated in patients with NSCLC, dynamic changes in serum NSE levels are controversial in predicting the efficacy of immunotherapy. Bello et al. reported that dynamic monitoring of NSE levels could not predict survival in patients with NSCLC treated with immunotherapy [26]. However, Chen et al. reported that dynamic monitoring of NSE levels could predict survival in patients with NSCLC treated with immunotherapy [24]. Both studies included a small number of patients, did not differentiate pathological subtypes, and included less than 50% of the patients who received first-line immunotherapy [24, 26]. Therefore, there may have been potential bias. Baseline NSE levels predict survival in patients with advanced NSCLC treated with targeted therapy. However, no studies have confirmed an association between baseline NSE levels and immunotherapy efficacy. In our study, baseline NSE levels predicted first-line immunotherapy efficacy in patients with LUAD and LUSC; the higher the baseline NSE levels, the inferior the PFS and OS periods. We found that the serum baseline NSE levels and PD-L1 expression were independent of each other. Therefore, these levels can be used together with PD-L1 expression as an indicator of efficacy and survival prediction in future clinical practice.
CYFRA21-1 has also been widely used as a tumor marker for NSCLC in recent years [19, 20]. We found that, similar to NSE, it predicted PFS and OS with first-line immunotherapy, except that it did not lead to inferior PFS with higher concentrations of CYFRA21-1 at baseline in patients with LUAD.
CA19-9 is mainly used in digestive tract tumors but is also elevated in NSCLC [23]. We found that in patients with LUAD, baseline CA19-9 level remained a valid predictor, and a higher CA19-9 level was associated with inferior OS. However, in patients with LUSC, CA19-9 level was not an independent prognostic factor.
CA125 is a classic tumor marker in ovarian cancer [21], but more than half of the patients with advanced NSCLC have CA125 levels higher than the upper limit of the normal value. In our study, CA125, similar to NSE, can effectively predict patients who can benefit from immunotherapy, and the higher the baseline CA125 level, the inferior the PFS and OS periods.
STMs can also serve as powerful complements to the treatment response in patients with LUAD. The baseline levels of CEA, NSE, and CA19-9 were statistically different from those of ORR and DCR. However, this phenomenon is not evident in patients with LUSC, suggesting the complexity of immunotherapy treatment response and efficacy prediction.
Our study has some limitations. First, this retrospective study had some selection bias. Second, there was no dynamic monitoring of STMs, and it was difficult to comprehensively evaluate the value of STMs in immunotherapy. Third, lung cancer is a highly heterogeneous malignant tumor, and whether the increase in NSE was due to a combination of small cells was not determined in this study.
To the best of our knowledge, this is the largest multicenter retrospective study to investigate the efficacy of baseline STMs in first-line immunotherapy. We further subdivided the patients into three groups based on STM levels and further subdivided the patients who could benefit from immunotherapy. It was also confirmed that the level of certain STMs at baseline affects immunotherapy efficacy rather than simply dichotomizing STMs based on the upper limit of the normal value. In addition, we found that PD-L1 expression levels were independent of most STM levels. Therefore, STMs can be used as effective prognostic factors in addition to PD-L1 in subsequent clinical applications. Finally, STMs routinely measured in clinics is an easy and effective tool to predict the efficacy of immunotherapy.

Conclusion

In patients with LUAD, STMs predict PFS and OS with first-line immunotherapy; higher serum NSE, CYFRA21-1, and CA125 levels are associated with inferior PFS. In addition, higher serum NSE, CYFRA21-1, CA19-9, and CA125 levels are associated with inferior OS. In patients with LUSC, serum CEA, NSE, CYFRA21-1, and CA125 levels predict PFS and OS with first-line immunotherapy, and higher serum NSE, CYFRA21-1, and CA125 levels are associated with inferior PFS and OS. These findings need to be validated in large prospective studies.

Acknowledgements

We would like to thank Editage (www.​editage.​cn) for English language editing.

Declarations

All procedures involving collection of tissue were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. This study was approved by the ethics committee of Jiangxi Cancer Hospital, the Third Affiliated Hospital of Sun Yat sen University, General Hospital of Southern Theater Command and the Third Xiangya Hospital of Central South University. Written informed consent was obtained from individual or guardian participants.
Not applicable.

Competing interests

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

Publisher’s Note

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

Supplementary Information

Literatur
2.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021 n/a(n/a). Epub 2021/02/05. https://doi.org/10.3322/caac.21660. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021 n/a(n/a). Epub 2021/02/05.  https://​doi.​org/​10.​3322/​caac.​21660.
3.
Zurück zum Zitat Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378(22):2078–92.CrossRefPubMed Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378(22):2078–92.CrossRefPubMed
4.
Zurück zum Zitat Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379(21):2040–51.CrossRefPubMed Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379(21):2040–51.CrossRefPubMed
5.
Zurück zum Zitat Gadgeel S, Rodriguez-Abreu D, Speranza G, Esteban E, Felip E, Domine M, et al. Updated analysis from keynote-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer. J Clin Oncol. 2020;8(14):1505–17.CrossRef Gadgeel S, Rodriguez-Abreu D, Speranza G, Esteban E, Felip E, Domine M, et al. Updated analysis from keynote-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer. J Clin Oncol. 2020;8(14):1505–17.CrossRef
6.
Zurück zum Zitat Rodríguez-Abreu D, Powell SF, Hochmair MJ, Gadgeel S, Esteban E, Felip E, et al. Pemetrexed plus platinum with or without pembrolizumab in patients with previously untreated metastatic nonsquamous nsclc: protocol-specified final analysis from keynote-189. Ann Oncol. 2021;32(7):881–95.CrossRefPubMed Rodríguez-Abreu D, Powell SF, Hochmair MJ, Gadgeel S, Esteban E, Felip E, et al. Pemetrexed plus platinum with or without pembrolizumab in patients with previously untreated metastatic nonsquamous nsclc: protocol-specified final analysis from keynote-189. Ann Oncol. 2021;32(7):881–95.CrossRefPubMed
7.
Zurück zum Zitat Suresh K, Naidoo J, Lin CT, Danoff S. Immune checkpoint immunotherapy for non-small cell lung cancer: benefits and pulmonary toxicities. Chest. 2018;154(6):1416–23.CrossRefPubMedPubMedCentral Suresh K, Naidoo J, Lin CT, Danoff S. Immune checkpoint immunotherapy for non-small cell lung cancer: benefits and pulmonary toxicities. Chest. 2018;154(6):1416–23.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Liu L, Bai H, Wang C, Seery S, Wang Z, Duan J, et al. Efficacy and safety of first-line immunotherapy combinations for advanced nsclc: a systematic review and network meta-analysis. J Thorac Oncol. 2021;16(7):1099–117.CrossRefPubMed Liu L, Bai H, Wang C, Seery S, Wang Z, Duan J, et al. Efficacy and safety of first-line immunotherapy combinations for advanced nsclc: a systematic review and network meta-analysis. J Thorac Oncol. 2021;16(7):1099–117.CrossRefPubMed
9.
Zurück zum Zitat Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for pd-l1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823–33.CrossRefPubMed Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for pd-l1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823–33.CrossRefPubMed
10.
Zurück zum Zitat Ricciuti B, Wang X, Alessi JV, Rizvi H, Mahadevan NR, Li YY, et al. Association of high tumor mutation burden in non-small cell lung cancers with increased immune infiltration and improved clinical outcomes of Pd-L1 blockade across Pd-L1 expression levels. JAMA Oncol. 2022;8(8):1160–8.CrossRefPubMedPubMedCentral Ricciuti B, Wang X, Alessi JV, Rizvi H, Mahadevan NR, Li YY, et al. Association of high tumor mutation burden in non-small cell lung cancers with increased immune infiltration and improved clinical outcomes of Pd-L1 blockade across Pd-L1 expression levels. JAMA Oncol. 2022;8(8):1160–8.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Stadler JC, Belloum Y, Deitert B, Sementsov M, Heidrich I, Gebhardt C, et al. Current and future clinical applications of ctdna in immuno-oncology. Cancer Res. 2022;82(3):349–58.CrossRefPubMed Stadler JC, Belloum Y, Deitert B, Sementsov M, Heidrich I, Gebhardt C, et al. Current and future clinical applications of ctdna in immuno-oncology. Cancer Res. 2022;82(3):349–58.CrossRefPubMed
12.
Zurück zum Zitat Fiala O, Pesek M, Finek J, Svaton M, Sorejs O, Bortlicek Z, et al. Prognostic significance of serum tumor markers in patients with advanced-stage nsclc treated with pemetrexed-based chemotherapy. Anticancer Res. 2016;36(1):461–6 (Epub 2016/01/02).PubMed Fiala O, Pesek M, Finek J, Svaton M, Sorejs O, Bortlicek Z, et al. Prognostic significance of serum tumor markers in patients with advanced-stage nsclc treated with pemetrexed-based chemotherapy. Anticancer Res. 2016;36(1):461–6 (Epub 2016/01/02).PubMed
13.
Zurück zum Zitat Kulda V, Hrda K, Houdek Z, Dobra JK, Vrzakova R, Svaton M, et al. Predictive significance of thymidylate synthase expression in non-small cell lung cancer. Anticancer Res. 2017;37(12):6953–8.PubMed Kulda V, Hrda K, Houdek Z, Dobra JK, Vrzakova R, Svaton M, et al. Predictive significance of thymidylate synthase expression in non-small cell lung cancer. Anticancer Res. 2017;37(12):6953–8.PubMed
14.
Zurück zum Zitat Baek AR, Seo HJ, Lee JH, Park SW, Jang AS, Paik SH, et al. Prognostic Value of baseline carcinoembryonic antigen and cytokeratin 19 fragment levels in advanced non-small cell lung cancer. Cancer Biomark. 2018;22(1):55–62.CrossRefPubMed Baek AR, Seo HJ, Lee JH, Park SW, Jang AS, Paik SH, et al. Prognostic Value of baseline carcinoembryonic antigen and cytokeratin 19 fragment levels in advanced non-small cell lung cancer. Cancer Biomark. 2018;22(1):55–62.CrossRefPubMed
15.
Zurück zum Zitat Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WE, et al. The Iaslc lung cancer staging project: proposals for revision of the tnm stage groupings in the forthcoming (Eighth) edition of the tnm classification for lung cancer. J Thorac Oncol. 2016;11(1):39–51.CrossRefPubMed Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WE, et al. The Iaslc lung cancer staging project: proposals for revision of the tnm stage groupings in the forthcoming (Eighth) edition of the tnm classification for lung cancer. J Thorac Oncol. 2016;11(1):39–51.CrossRefPubMed
16.
Zurück zum Zitat Board WCoTE. Who Classification of Tumours. Thoracic Tumours. IARC Publications 2021. Board WCoTE. Who Classification of Tumours. Thoracic Tumours. IARC Publications 2021.
17.
Zurück zum Zitat Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised recist guideline (Version 1.1). Eur J Cancer. 2009;45(2):228–47.CrossRefPubMed Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised recist guideline (Version 1.1). Eur J Cancer. 2009;45(2):228–47.CrossRefPubMed
18.
Zurück zum Zitat Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, Pd-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (Keynote-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019;393(10183):1819–30.CrossRefPubMed Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, Pd-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (Keynote-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019;393(10183):1819–30.CrossRefPubMed
19.
Zurück zum Zitat Ardizzoni A, Cafferata MA, Tiseo M, Filiberti R, Marroni P, Grossi F, et al. Decline in serum carcinoembryonic antigen and cytokeratin 19 fragment during chemotherapy predicts objective response and survival in patients with advanced nonsmall cell lung cancer. Cancer. 2006;107(12):2842–9.CrossRefPubMed Ardizzoni A, Cafferata MA, Tiseo M, Filiberti R, Marroni P, Grossi F, et al. Decline in serum carcinoembryonic antigen and cytokeratin 19 fragment during chemotherapy predicts objective response and survival in patients with advanced nonsmall cell lung cancer. Cancer. 2006;107(12):2842–9.CrossRefPubMed
20.
Zurück zum Zitat Yang L, Chen X, Li Y, Yang J, Tang L. Declines in Serum Cyfra21–1 and Carcinoembryonic antigen as predictors of chemotherapy response and survival in patients with advanced non-small cell lung cancer. Exp Ther Med. 2012;4(2):243–8.CrossRefPubMed Yang L, Chen X, Li Y, Yang J, Tang L. Declines in Serum Cyfra21–1 and Carcinoembryonic antigen as predictors of chemotherapy response and survival in patients with advanced non-small cell lung cancer. Exp Ther Med. 2012;4(2):243–8.CrossRefPubMed
21.
Zurück zum Zitat Bast RC Jr, Badgwell D, Lu Z, Marquez R, Rosen D, Liu J, et al. New Tumor Markers: Ca125 and Beyond. Int J Gynecol Cancer. 2005;15(Suppl 3):274–81.CrossRefPubMed Bast RC Jr, Badgwell D, Lu Z, Marquez R, Rosen D, Liu J, et al. New Tumor Markers: Ca125 and Beyond. Int J Gynecol Cancer. 2005;15(Suppl 3):274–81.CrossRefPubMed
22.
Zurück zum Zitat de Kock R, Borne BVD, Soud MY, Belderbos H, Stege G, de Saegher M, et al. Circulating biomarkers for monitoring therapy response and detection of disease progression in lung cancer patients. Cancer Treat Res Commun. 2021;28:100410.CrossRefPubMed de Kock R, Borne BVD, Soud MY, Belderbos H, Stege G, de Saegher M, et al. Circulating biomarkers for monitoring therapy response and detection of disease progression in lung cancer patients. Cancer Treat Res Commun. 2021;28:100410.CrossRefPubMed
23.
Zurück zum Zitat Scarà S, Bottoni P, Scatena R. Ca 19–9: Biochemical and Clinical Aspects. Adv Exp Med Biol. 2015;867:247–60.CrossRefPubMed Scarà S, Bottoni P, Scatena R. Ca 19–9: Biochemical and Clinical Aspects. Adv Exp Med Biol. 2015;867:247–60.CrossRefPubMed
24.
Zurück zum Zitat Chen Y, Wen S, Xia J, Du X, Wu Y, Pan B, et al. Association of dynamic changes in peripheral blood indexes with response to Pd-1 inhibitor-based combination therapy and survival among patients with advanced non-small cell lung cancer. Front Immunol. 2021;12:672271.CrossRefPubMedPubMedCentral Chen Y, Wen S, Xia J, Du X, Wu Y, Pan B, et al. Association of dynamic changes in peripheral blood indexes with response to Pd-1 inhibitor-based combination therapy and survival among patients with advanced non-small cell lung cancer. Front Immunol. 2021;12:672271.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Zhang Z, Yuan F, Chen R, Li Y, Ma J, Yan X, et al. Dynamics of serum tumor markers can serve as a prognostic biomarker for chinese advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors. Front Immunol. 2020;11:1173.CrossRefPubMedPubMedCentral Zhang Z, Yuan F, Chen R, Li Y, Ma J, Yan X, et al. Dynamics of serum tumor markers can serve as a prognostic biomarker for chinese advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors. Front Immunol. 2020;11:1173.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Dal Bello MG, Filiberti RA, Alama A, Orengo AM, Mussap M, Coco S, et al. The Role of Cea, Cyfra21–1 and Nse in monitoring tumor response to nivolumab in advanced non-small cell lung cancer (Nsclc) Patients. J Transl Med. 2019;17(1):74.CrossRef Dal Bello MG, Filiberti RA, Alama A, Orengo AM, Mussap M, Coco S, et al. The Role of Cea, Cyfra21–1 and Nse in monitoring tumor response to nivolumab in advanced non-small cell lung cancer (Nsclc) Patients. J Transl Med. 2019;17(1):74.CrossRef
27.
Zurück zum Zitat Lang D, Horner A, Brehm E, Akbari K, Hergan B, Langer K, et al. Early serum tumor marker dynamics predict progression-free and overall survival in single Pd-1/Pd-L1 inhibitor treated advanced Nsclc-a retrospective cohort study. Lung Cancer. 2019;134:59–65.CrossRefPubMed Lang D, Horner A, Brehm E, Akbari K, Hergan B, Langer K, et al. Early serum tumor marker dynamics predict progression-free and overall survival in single Pd-1/Pd-L1 inhibitor treated advanced Nsclc-a retrospective cohort study. Lung Cancer. 2019;134:59–65.CrossRefPubMed
28.
Zurück zum Zitat Grunnet M, Sorensen JB. Carcinoembryonic Antigen (Cea) as Tumor Marker in Lung Cancer. Lung Cancer. 2012;76(2):138–43.CrossRefPubMed Grunnet M, Sorensen JB. Carcinoembryonic Antigen (Cea) as Tumor Marker in Lung Cancer. Lung Cancer. 2012;76(2):138–43.CrossRefPubMed
29.
Zurück zum Zitat Kasprzak A, Zabel M, Biczysko W. Selected Markers (Chromogranin a, Neuron-Specific Enolase, Synaptophysin, Protein Gene Product 9.5) in diagnosis and prognosis of neuroendocrine pulmonary tumours. Pol J Pathol. 2007;58(1):23–33.PubMed Kasprzak A, Zabel M, Biczysko W. Selected Markers (Chromogranin a, Neuron-Specific Enolase, Synaptophysin, Protein Gene Product 9.5) in diagnosis and prognosis of neuroendocrine pulmonary tumours. Pol J Pathol. 2007;58(1):23–33.PubMed
Metadaten
Titel
Baseline serum tumor markers predict the survival of patients with advanced non-small cell lung cancer receiving first-line immunotherapy: a multicenter retrospective study
verfasst von
Jian Huang
Yi Xiao
Yubin Zhou
Huiyin Deng
Zihao Yuan
Longyan Dong
Jun Lan
Xiane Li
Gaijiao Liu
Hao Hu
Shaohong Huang
Xiongwen Yang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11312-4

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

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

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

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