Abstract
Background
Neoadjuvant immunotherapy has been demonstrated to be effective and safe in resectable non-small cell lung cancer (NSCLC) patients. However, the presence of different oncogenic driver mutations may affect the tumor microenvironment and consequently influence the clinical benefit from immunotherapy.
Methods
This retrospective study included consecutive NSCLC patients (stage IIA to IIIB) who underwent radical surgery after receiving neoadjuvant immunotherapy at a single high-volume center between December 2019 and August 2022. Pathological response and long-term outcomes were compared based on the driver oncogene status, and RNA sequencing analysis was conducted to investigate the transcriptomic characteristics before and after treatment.
Results
Of the 167 patients included in this study, 47 had oncogenic driver mutations. KRAS driver mutations were identified in 28 patients, representing 59.6% of oncogenic driver mutations. Of these, 17 patients had a major pathological response, which was significantly higher than in the non-KRAS driver mutation group (60.7% vs. 31.6%, P = 0.049). Multivariate Cox regression analysis further revealed that the KRAS driver mutation group was an independent prognostic factor for prolonged disease-free survival (hazard ratio: 0.10, P = 0.032). The median proportion of CD8+ T cells was significantly higher in the KRAS driver mutation NSCLCs than in the non-driver mutation group (18% vs. 13%, P = 0.030). Furthermore, immune-related pathways were enriched in the KRAS driver mutation NSCLCs and activated after immunotherapy.
Conclusion
Our study suggests that NSCLC patients with KRAS driver mutations have a superior response to neoadjuvant immunotherapy, possibly due to their higher immunogenicity. The findings highlight the importance of considering oncogenic driver mutations in selecting neoadjuvant treatment strategies for NSCLC patients.
Supplementary Information
The online version contains supplementary material available at 10.1007/s00262-023-03560-x.
Keywords: Neoadjuvant immunotherapy, Immune checkpoint inhibitors, Non-small cell lung cancer, Oncogenic driver mutations, KRAS driver mutations
Introduction
Lung cancer remains the foremost cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all lung cancer cases [1]. NSCLC has emerged as a paradigm of precision medicine in recent decades due to the identification of various disease subtypes that are defined by specific oncogenic driver mutations, leading to the development of a range of molecularly targeted therapies [2]. Simultaneously, significant strides have been made in the development of immune-checkpoint inhibitors (ICIs), particularly antagonistic antibodies that target the PD-1/PD-L1 axis, for treatment of numerous types of cancers including NSCLC [3, 4].
For decades, chemotherapy has remained the standard neoadjuvant treatment for resectable NSCLCs [5]. Recently, neoadjuvant therapy with nivolumab in combination with platinum-based chemotherapy was approved by the U.S. Food and Drug Administration as the first-line treatment for NSCLC patients, with pembrolizumab also being added to preoperative neoadjuvant indications in the latest National Comprehensive Cancer Network (NCCN) guideline [6–8]. Although the CheckMate 816 study showed encouraging results for the combination of immunotherapy with chemotherapy in patients with resectable stage IB to IIIA NSCLC [6], it remains unclear whether different driver mutation subtypes influence the response to immunotherapy [9]. Some recent studies found that oncogene-positive NSCLC respond less than oncogene-negative NSCLC following neoadjuvant immunotherapy [10, 11], while other study demonstrated the opposite results [12].
The efficacy of ICIs in NSCLC is based on the immunogenicity of tumors and corresponds with the expression of PD-L1 on tumor cells, tumor mutational burden (TMB), and the immune infiltration within the tumor microenvironment [13]. Different oncogenic driver mutations in NSCLC are associated with distinct tumor immune microenvironments, which are closely linked to the efficacy of ICIs [14]. Although patients with specific oncogenic driver mutations in advanced NSCLC have been recognized as poorly responsive to immunotherapy, evidence in the resectable stage or locally advanced setting is still lacking [15, 16]. Furthermore, the efficacy of neoadjuvant immunotherapy on long-term survival in patients with different oncogenic driver alterations remains to be explored.
Therefore, the aim of this study is to investigate the impact of oncogenic driver mutations in patients with resectable NSCLC who underwent radical surgery after the completion of neoadjuvant immunotherapy. Through further RNA sequencing analysis, we aim to provide insights into the potential mechanisms that lead to differential response of neoadjuvant immunotherapy.
Methods
Study design
This study was a retrospective, single-center study. Eligible patients were defined as: 1. treated with neoadjuvant immunotherapy (including PD-1 inhibitors and PD-L1 inhibitors); 2. diagnosed with IIA to IIIB NSCLC, as determined by the American Joint Committee on Cancer (AJCC, 8th edition); 3. received radical surgery at Shanghai Pulmonary Hospital after the completion of neoadjuvant immunotherapy between December 2019 and August 2022. The exclusion criteria of patients were as follows: 1. incomplete treatment modality or clinical information; 2. M1 metastases or N3 lymph node metastases; 3. pathologically confirmed locoregional relapses; 4. histologically confirmed small cell lung cancer (SCLC) or SCLC combined with NSCLC; 5. targeted therapy prior to the radical surgery; 6. without gene testing. The flowchart of this study is shown in Fig. 1. The enrolled patients were divided into driver mutation group and non-driver mutation group. Due to KRAS mutations being the most prevalent oncogenic driver mutations in the patients who received immunotherapy in this study, driver mutation group was further subdivided into KRAS driver mutations and non-KRAS driver mutations to analyze short-term outcomes and long-term survival among the three groups.
Fig. 1.
Study flowchart. SCLC, small cell lung cancer
This study was conducted in accordance with the Declaration of Helsinki and was approved by the institutional review board of Shanghai Pulmonary Hospital (K23-203). Informed consent was waived due to the retrospective and observational nature of the study. Only patients with telephone follow-up were provided with informed consent in this study.
Data collection and follow-up
The demographic and clinical data of the included patients, including clinical characteristics, molecular profiles, treatment details, surgical parameters, pathological data, and survival information, were collected retrospectively. The KRAS, EGFR, BRAF, ROS1, and ALK status of all enrolled patients were tested, while other oncogene (RET, HER-2 and c-MET) status was tested in part of the NSCLC patients through next-generation sequencing, immunohistochemistry (IHC), or polymerase chain reaction (PCR) by using tumor samples. In accordance with the manufacturer’s instructions, the majority of patients (94.6%) in this study underwent Amplification Refractory Mutation System-qPCR method for hotspot mutation detection using the genetic mutation assay kit from Beijing ACCB Biotech Ltd. The panel of PCR mainly consisted of two types: (1) KRAS, EGFR, BRAF, ROS1, ALK; (2) KRAS, EGFR, BRAF, ROS1, ALK, RET, NRAS, PIK3CA, HER-2 and C-MET. In addition, IHC was conducted in some patients to detect ALK fusions using the VENTANA D5F3 antibody clone. The oncogenic driver mutations were defined as any of the following events: KRAS mutations, EGFR mutations, HER-2 amplifications or mutations, BRAF fusions or mutations, RET fusions, ALK translocations or mutations, ROS1 rearrangements or mutations, or C-MET amplifications or mutations.
PD-L1 expression was quantified as the proportion of PD-L1 positive tumor cells through PD-L1 immunohistochemistry assay. Cases with < 100 total tumor cells for scoring were defined as not applicable (NA). The short-term outcomes of interest were rates of major pathologic response (MPR) and pathologic complete response (pCR). MPR was defined as the presence of 10% or fewer residual viable tumor cells, while pCR was defined as no viable tumor cells in both primary and lymph nodes.
Follow-up information is updated periodically through clinics visit, and telephone follow-up was conducted if outpatient follow-up was not feasible through medical records. The end point of follow-up for this study was January 31, 2023.
The long-term outcomes were also analyzed according to the driver mutation status, including overall survival (OS) and disease-free survival (DFS). OS of patients was calculated as the duration between radical surgery and death from any cause. DFS was calculated as the period between the initial surgery and the first occurrence of either tumor recurrence, metastasis, or death from any cause. Survival data were censored at the date of last follow-up.
RNA sequencing analysis
The RNA-seq data of 12 baseline tumor samples and 26 surgical tumor samples were partially obtained from 30 NSCLC patients who were recruited in two published phase II studies of neoadjuvant anti-PD-1 therapy at Shanghai Pulmonary Hospital (LungMate 001 and LungMate 002) [17, 18]. The protocol of sample preparation, library preparation and sequencing were conducted as described previously [18].
The pathway analyses were performed through gene set enrichment analysis (GSEA) with the downloaded GSEA software (www.broadinstitute.org/gsea). The GSEA results based on the criterion of P value < 0.05 and false discovery rate (FDR) ≤ 0.25 were filtered. Then, candidate pathways were visualized based on the normalized enrichment score from the filtered list.
The proportion of the immune cell infiltration of each sample was evaluated by cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm based on the LM22 gene set [19]. The relevant procedure was carried out using CIBERSORT package in R.
Statistical analysis
The analysis of categorical variables was carried out through either the chi-square test or Fisher's exact test, as appropriate. For continuous variables with normal distribution, the mean and standard deviation (SD) were calculated and analyzed using Student's t test. Conversely, for continuous variables with non-normal distribution, the median and interquartile range (IQR) were computed and analyzed using the Kruskal–Wallis test. The survival analysis of OS and DFS across mutation groups was conducted using the log-rank test and was visually represented through the Kaplan–Meier curve. To assess the association between each variable and survival outcomes, hazard ratios (HR) and their confidence intervals (CI) were calculated through Cox proportional hazards regression models.
The statistical analyses were conducted using R version 4.2.2. All tests were two-tailed, and P < 0.05 was considered statistically significant.
Results
Clinical characteristics of patients
This study involved a retrospective screening of 241 patients who underwent radical surgery subsequent to receiving neoadjuvant immunotherapy, spanning from December 2019 to August 2022. The exclusion criteria were duly taken into account, resulting in a final sample size of 167 patients for analysis (Fig. 1). The mean age of the study population was 62 years, with males comprising 89.8% of the cohort, and 95.2% (159/167) of the patients received immunotherapy combined with chemotherapy. Among the cohort of enrolled patients, 47 (28.1%) patients presented with driver mutations. Comparison of baseline clinical characteristics between the driver mutation group and the non-driver mutation group revealed no statistically significant distinctions (Supplementary table 1).
To investigate the proportion of driver gene mutations in the driver mutation group, we visualize the detailed information of driver mutation group by waterfall plot (Fig. 2A). Among them, there were 59.6% (28/47) were found to have KRAS mutations, while 17.0% (8/47) harbored EGFR mutations (Fig. 2B). Despite NSCLC patients with EGFR mutations often serving as contraindications for immunotherapy, several patients with EGFR mutations received neoadjuvant immunotherapy in this cohort due to insufficient tissue samples or false negative biopsy results, while further genetic testing of surgical specimens revealed the presence of EGFR mutations. Other oncogenemutations, such as RET fusion, ROS1 fusion, BRAF, C-MET, and HER-2 mutations, accounted for 23.4% (11/47) of driver mutations in this cohort. Detailed information of patients with non-KRAS driver mutations was shown in Supplementary table 2. The percentage of residual viable tumor cells displayed heterogeneity among the various oncogenic driver mutations, and accordingly, the MPR and pCR rates also varied (Supplementary Table 3). Given that KRAS driver mutations constituted the majority of driver mutations, and the residual viable tumor cells of KRAS driver mutations were significantly lower than the other driver mutations (2.5% vs. 30.0%, P = 0.014, Fig. 2C), the driver mutation group was divided into two groups for further analysis: the KRAS driver mutations and the non-KRAS driver mutations.
Fig. 2.
Overview of the clinicopathological characteristics of neoadjuvant immunotherapy-treated NSCLCs with oncogenic driver mutations. A Waterfall plot of clinicopathological characteristics of patients with resected NSCLC. B The pie chart shows the distribution and proportion of each oncogenic driver mutations. C Comparison of the residual viable tumor cells between the KRAS driver mutation group and the non-KRAS driver mutation group. LUAD lung adenocarcinoma; LUSC lung squamous cell carcinoma; MPR major pathological response; pCR pathological complete response
Among all patients, the non-KRAS driver mutation group consisted of a higher proportion of females and never smokers compared to the other two groups (Table 1). Furthermore, non-KRAS driver mutation group had the highest ratio of forced expiratory volume in 1 s/forced vital capacity (FEV1/FVC), with a median of 102.30% among the three groups (P = 0.020), this may be related to the fact that smokers have the lowest proportion among this group of patients. Other characteristics including age, body mass index (BMI), Eastern Cooperative Oncology Group (ECOG) score, driver mutation detective method, stage at diagnosis and neoadjuvant regimes were comparable among the three groups.
Table 1.
Baseline clinical characteristics of included patients stratified by driver mutations
Variables | KRAS driver mutations | Non-KRAS driver mutations | Non-driver mutations | P-value |
---|---|---|---|---|
Number | 28 | 19 | 120 | |
Age, years (mean (SD)) | 63.89 (8.26) | 59.79 (9.03) | 61.98 (7.81) | 0.227 |
Gender (%) | ||||
Male | 27 (96.4) | 12 (63.2) | 111 (92.5) | < 0.001 |
Female | 1 (3.6) | 7 (36.8) | 9 (7.5) | |
BMI, kg/m2 (mean (SD)) | 22.75 (3.40) | 23.65 (0.82) | 23.40 (2.45) | 0.768 |
ECOG score (%) | ||||
0 | 17 (60.7) | 13 (68.4) | 80 (66.7) | 0.811 |
1 | 11 (39.3) | 6 (31.6) | 40 (33.3) | |
Smoking status (%) | ||||
Never | 6 (21.4) | 10 (52.6) | 31 (25.8) | 0.023 |
Former smoker | 2 (7.1) | 3 (15.8) | 6 (5.0) | |
Current smoker | 20 (71.4) | 6 (31.6) | 83 (69.2) | |
FEV1/FVC, % (median [IQR]) | 93.85 [84.28, 97.00] | 102.30 [94.60, 104.10] | 95.65 [90.67, 101.05] | 0.020 |
Detective method (%) | ||||
NGS | 2 (7.1) | 1 (5.3) | 6 (5.0) | 0.903 |
PCR | 26 (92.9) | 18 (94.7) | 114 (95.0) | |
Stage at diagnosis (%) | ||||
IIA/IIB | 5 (17.9) | 1 (5.3) | 20 (16.7) | 0.415 |
IIIA/IIIB | 23 (82.1) | 18 (94.7) | 100 (83.3) | |
Neoadjuvant regimens (%) | ||||
ICI alone | 1 (3.6) | 0 (0.0) | 7 (5.8) | 0.513 |
ICI + chemotherapy | 27 (96.4) | 19 (100.0) | 113 (94.2) |
SD standard deviation; BMI body mass index; ECOG Eastern Cooperative Oncology Group; FEV1 forced expiratory volume in 1 s; FVC forced vital capacity; IQR interquartile range; PCR polymerase chain reaction; NGS next-generation sequencing; ICI immune checkpoint inhibitor
All patients with driver mutations received PD-1 inhibitors as neoadjuvant settings, while 11.7% of the patients in non-driver mutation group received PD-L1 inhibitors (Supplementary Table 4). Carboplatin-based and cisplatin-based chemotherapy were used in patients with combination therapy. The neoadjuvant cycles were well-balanced among the three groups (P = 0.875), and the rate of postoperative immunotherapy or postoperative chemotherapy showed no significance among the three groups (P = 0.497 and P = 0.551, respectively).
Surgical profile and pathological information
The majority of patients (80.8%) underwent video-assisted thoracoscopic surgery (VATS), with lobectomy being the most commonly used resection method (Table 2). Only one patient had positive margins. The surgical approach, resection method, operative time, estimated blood loss and resection margins were comparable among the three groups. As shown in Table 3, the tumor size and staging parameters showed no significance among the three groups. However, the histology was found to be different among the three groups (P < 0.001), with the KRAS driver mutation group having the highest proportion of lung adenocarcinoma (LUAD) (78.6%), while patients in the non-driver mutation group were more likely to be diagnosed with lung squamous cell carcinoma (LUSC) (53.3%). Furthermore, the KRAS driver mutation group had the lowest residual viable tumor cells (Supplementary Fig. 1A), and highest MPR rate and pCR rate among the three groups (Supplementary Fig. 1B). Specifically, the MPR rate of the KRAS driver mutation group was significantly higher than the non-KRAS driver mutation group (60.7% vs. 31.6%, P = 0.049).
Table 2.
Intraoperative parameters of patients who received immunotherapy stratified by driver mutations
Variables | KRAS driver mutations | Non-KRAS driver mutations | Non-driver mutations | P-value |
---|---|---|---|---|
Number | 28 | 19 | 120 | |
Surgical approach (%) | ||||
VATS | 22 (78.6) | 16 (84.2) | 97 (80.8) | 0.890 |
Open surgery | 6 (21.4) | 3 (15.8) | 23 (19.2) | |
Resection method (%) | ||||
Pneumonectomy | 1 (3.6) | 3 (15.8) | 5 (4.2) | 0.102 |
Lobectomy | 27 (96.4) | 16 (84.2) | 115 (95.8) | |
Operative time, min (median [IQR]) | 139.50 [129.75, 199.75] | 140.00 [105.50, 191.00] | 164.00 [122.50, 199.00] | 0.743 |
Estimated blood loss, ml (median [IQR]) | 50.00 [50.00, 100.00] | 50.00 [50.00, 100.00] | 50.00 [50.00, 100.00] | 0.973 |
Resection margins (%) | ||||
R0 | 28 (100.0) | 19 (100.0) | 119 (99.2) | 0.821 |
R1/2 | 0 (0.0) | 0 (0.0) | 1 (0.8) |
VATS video-assisted thoracoscopic surgery; IQR interquartile range
Table 3.
Pathological data of patients who received immunotherapy stratified by driver mutations
Variables | KRAS driver mutations | Non-KRAS driver mutations | Non-driver mutations | P-value |
---|---|---|---|---|
Number | 28 | 19 | 120 | |
Tumor size, mm (median [IQR]) | 30.50 [20.00, 39.25] | 32.00 [23.50, 36.00] | 26.50 [19.75, 39.25] | 0.695 |
pT stage (%) | ||||
T1 | 14 (50.0) | 8 (42.1) | 70 (58.3) | 0.185 |
T2 | 10 (35.7) | 10 (52.6) | 38 (31.7) | |
T3 | 2 (7.1) | 1 (5.3) | 11 (9.2) | |
T4 | 2 (7.1) | 0 (0.0) | 1 (0.8) | |
pN stage (%) | ||||
N0 | 19 (67.9) | 10 (52.6) | 66 (55.0) | 0.587 |
N1 | 2 (7.1) | 4 (21.1) | 16 (13.3) | |
N2 | 7 (25.0) | 5 (26.3) | 38 (31.7) | |
Histology (%) | ||||
LUAD | 22 (78.6) | 13 (68.4) | 43 (35.8) | < 0.001 |
LUSC | 2 (7.1) | 3 (15.8) | 64 (53.3) | |
Others | 4 (14.3) | 3 (15.8) | 13 (10.8) | |
MPR (%) | 17 (60.7) | 6 (31.6) | 58 (48.3) | 0.146 |
pCR (%) | 10 (35.7) | 2 (10.5) | 27 (22.5) | 0.123 |
IQR interquartile range; LUAD lung adenocarcinoma; LUSC lung squamous cell carcinoma; MPR major pathological response; pCR pathological complete response
Long-term outcomes
To investigate whether the driver mutation status has a long-term impact on patients who received neoadjuvant immunotherapy, a survival analysis based on DFS and OS was performed. In the entire cohort, the 1-year recurrence rate and 1-year survival rate were 88.2% and 96.1%, respectively, with a median follow-up time of 13 months (IQR, 7–18 months). The KRAS driver mutation group was associated with the lowest rate of recurrence events and deaths among the three groups (Fig. 3A, B). Given the potential survival benefit of KRAS mutation group compared with non-driver mutation group, a subgroup analysis was performed between the two groups. Patients in the KRAS driver mutation group had a significantly higher DFS rate compared with patients in non-driver mutation group (P = 0.030) and a higher OS rate that approached significance (P = 0.092). On multivariate Cox regression analysis predicting DFS, KRAS driver mutations were found to be an independent favorable prognostic factor (HR: 0.10, CI: 0.01–0.82, P = 0.032) (Supplementary Table 5).
Fig. 3.
Survival analysis of the entire cohort. A Kaplan–Meier curves showing disease-free survival (DFS) of all patients stratified by driver oncogene status. B Kaplan–Meier curves showing overall survival (OS) of all patients stratified by driver oncogene status. C Comparison of DFS between the KRAS driver mutation group and the non-KRAS driver mutation group using Kaplan–Meier analysis. D Comparison of OS between the KRAS driver mutation group and the non-KRAS driver mutation group using Kaplan–Meier analysis
Transcriptomic analysis
Recent studies have emphasized that PD-L1 expression, TMB, immunogenicity, and CD8+ T cell tumor-infiltrating lymphocytes (TILs) are associated with the response to PD-1 blockade therapy in NSCLC patients [20]. A large pooled analysis has confirmed that PD-L1 expression and TMB were higher in KRAS driver mutation tumors than in KRAS wild-type tumors [21]. To further explore the differences in pathway enrichment before and after neoadjuvant immunotherapy based on the KRAS driver mutation status and investigate whether KRAS driver mutation NSCLC has a higher rate of CD8+ TILs, we performed transcriptomic analysis using RNA-seq data.
Considering the KRAS driver mutation group exhibits a higher survival benefit than the non-driver mutation group, and to eliminate potential confounding factors and account for the heterogenicity and small sample size within the non-KRAS driver mutation group, GSEA was performed only between the KRAS driver mutation group and the non-driver mutation group. Through ranked GSEA, we found that the top gene sets at baseline that showed a significant association with the KRAS mutations were involved in the antigen receptor mediated signaling pathway, adaptive immune response, and T cell activation pathways (Fig. 4A, B). This suggests that KRAS driver mutations were associated with higher immunogenicity, which are more likely to respond to immunotherapy. In contrast, the non-driver mutation group was associated with cell fate specification, DNA binding transcription activator activity, and DNA templated DNA replication. Furthermore, GSEA analysis of post-treatment tumor samples revealed immune-activated pathways including activation of immune response, positive regulation of immune response, regulation of T cell activation, which were enriched in KRAS driver mutation group, while DNA replication was still observed in non-driver mutation group (Fig. 4C, D).
Fig. 4.
Overview of gene set enrichment analysis (GSEA) between the KRAS driver mutation group and non-driver mutation group before and after neoadjuvant immunotherapy. A Bar plot illustrates the significant pathways enriched in the KRAS driver mutation group and non-driver mutation group before immunotherapy. B GSEA of baseline samples comparing KRAS driver mutation group and non-driver mutation group. C Bar plot depicts the significant pathways between KRAS driver mutation group and non-driver mutation group after immunotherapy. D GSEA of post-immunotherapy samples comparing KRAS driver mutation group and non-driver mutation group. The NES and FDR q-value are presented for each comparison. NES, normalized enrichment score; FDR, false discovery rate
To reveal the immune cell infiltration characteristics in the immune tumor microenvironment (TIME) between the KRAS driver mutation group and the non-driver mutation group, we computed 22 types of immune cell infiltration in different subgroups using the CIBERSORT algorithm (Fig. 5A). The median proportion of CD8+ T cell was significantly higher in KRAS driver mutation group at baseline (18% vs. 13%, P = 0.030). After treatment, the proportion of M2 macrophage increased in non-driver mutation group, but not in KRAS driver mutation group (16% vs. 26%, P = 0.044, Fig. 5B) while the proportion of M1 macrophage did not show difference both in KRAS driver mutation group and non-driver mutation group (P = 0.360 and P = 0.280, respectively, Fig. 5C). Notably, the M2 macrophages with high expression of CD163, CD204 and CD206 are induced by IL-4 and IL-13, which plays a key role in exerting immunomodulatory effects [22]. M2 macrophages could thus create a favorable immunosuppressive TME for tumor progression, which in turn could influence immunotherapy resistance and be associated with poor survival outcomes [23, 24]. However, the molecular biology mechanisms of macrophages in immunotherapy remain to be further explored.
Fig. 5.
Analysis of immune cell infiltration and characteristics reveal differences in the tumor immune microenvironment between KRAS driver mutation and non-driver mutation groups before and after immunotherapy. A The differences in immune cell infiltration between the KRAS driver mutation group and the non-driver mutation group before immunotherapy using CIBERSORT. B Box plot shows an increase in the proportion of M2 macrophages after immunotherapy in the non-driver mutation group. C Box plot displays the distribution of the proportion of M1 macrophages in the KRAS driver mutation group and the non-driver mutation group before and after immunotherapy
Discussion
The utilization of neoadjuvant chemotherapy in combination with immunotherapy, has become the prevailing standard of care for patients afflicted with resectable or locally advanced non-small cell lung cancer [8]. Nonetheless, the efficacy of neoadjuvant treatment may vary substantially due to the heterogeneity of NSCLC, particularly in terms of oncogenic driver mutations [14]. In this study, we observed that patients with KRAS driver mutations exhibited a higher rate of MPR compared to non-KRAS driver mutation group. Furthermore, the status of KRAS driver mutations has been identified as an independent favorable prognostic factor for DFS among the wider population of resectable NSCLC patients.
In the era of immuno-oncology, emerging evidence suggests that driver oncogenes have different effects on the TIME that influence the potential for clinical benefit from treatment with ICIs [14]. KRAS mutation is one of the major oncogenic driver mutations in NSCLCs, the rate of KRAS mutation in NSCLC patients was 27% in Caucasians [25], but lower in Asians (10%) [26]. The presence of KRAS mutation is prognostic of poor survival for patients with NSCLC when compared to the absence of KRAS mutation in early research [27]. Nevertheless, the efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutated NSCLC is still not well defined [15, 21, 28–31]. Several previous studies found KRAS mutation was associated with superior response in advanced NSCLC patients who received immunotherapy [15, 21], while other studies found KRAS mutation was not associated with prolonged survival even found KRAS was an independent adverse prognostic factor in immunotherapy [28–30]. These heterogeneous results may relate to the different stages of NSCLCs and different regime of ICIs. A recent systematic review demonstrated that the benefits of immunotherapy in early resectable lung cancer appeared to be better than those observed in advanced lung cancer, especially with the regard to the regimen of immunotherapy in combination with chemotherapy [32]. Nowadays, the indication of immunotherapy is becoming more and more widespread, and the combination of ICIs with chemotherapy has also become mainstream in the adjuvant immunotherapy. Thus, this study was designed to focus on the effect of oncogenic driver mutations on resectable NSCLCs.
Several studies have demonstrated that KRAS-mutant NSCLCs are correlated with an increasing proportion of PD-L1 expression, TMB and CD8+ TILs [21, 33]. According to further transcriptomic analysis of our dataset, the proportion of CD8+ TILs was significantly increased in the KRAS mutated NSCLCs, which is consistent with the previous study [21]. Additionally, KRAS mutation has been reported to promote inflammatory signaling in the early stages of tumor progression, and can cooperate with external factors such as tobacco or pollutants in the lung to create an inflammatory environment that enhances oncogenic signaling [34]. The higher proportion of current smokers in KRAS driver mutation group was also observed in this study. Furthermore, several immune-related pathways were observed in KRAS driver mutation NSCLCs and further activated after immunotherapy. This finding suggests that KRAS driver mutation is associated with higher immunogenicity and may facilitate a more favorable response to immunotherapy in resectable NSCLCs.
In a phase 1 trial involving 59 patients with metastatic non-small cell lung cancer, Sotorasib, a small molecule targeting KRASG12C, demonstrated promising anticancer activity and safety [35]. However, for KRAS-mutated NSCLC, chemotherapy with or without immunotherapy remained the standard care of neoadjuvant schema for the moment [8]. In contrast, NSCLCs with other oncogenic driver mutations have a more diverse array of targeted drugs available. Although EGFR mutations are often contraindicated for immunotherapy, eight patients with EGFR mutations in this cohort still received neoadjuvant immunotherapy due to insufficient tissue samples for genetic testing or false negative biopsy results, while further genetic testing of surgical specimens revealed the presence of EGFR mutations. Unsurprisingly, only one patient (12.5%) with EGFR L858R mutation achieved MPR after neoadjuvant immunotherapy, consistent with the research in the early exploratory phase, indicating that patients with EGFR mutations show limited responsiveness to ICIs [16, 36, 37]. Nevertheless, the EGFR-TKIs have exhibited satisfying efficacy and acceptable safety profile in neoadjuvant therapy [38, 39]. While ALK and ROS1 rearrangements are commonly linked with lower TMB [40], their response to ICIs is very limited according to several previous studies [15, 37]. Conversely, for ALK-positive NSCLC, several targeted treatments including crizotinib, alectinib have been approved and showed superior response than chemotherapy [41, 42]. Other driver mutations consisted of 21% oncogenic driver mutations in our dataset, the efficacy of ICIs in patients with those oncogenic driver mutations still require further exploration. The ongoing NAUTIKA1 study (NCT04302025) focuses on the effect of neoadjuvant targeted therapy, including alectinib, entrectinib, vemurafenib, cobimetinib, or pralsetinib, on untreated patients with resectable stage IB-IIIB NSCLC diagnosed with ALK, ROS1, NTRK, BRAF, or RET mutations. Other ongoing trials such as ALNEO (NCT05015010) and NeoADAURA (NCT04351555) trials are evaluating neoadjuvant targeted therapies regarding specific molecular genotyping.
It is crucial to acknowledge that the impact of oncogenic driver mutations in NSCLCs is not solely determined by the oncogene itself, but also by the concurrent genomic mutations that influence the molecular and clinical heterogeneity of NSCLC tumors [43]. In particular, co-occurring mutations in suppressor genes, such as TP53 and STK11, in KRAS-mutant NSCLCs have demonstrated significant implications for treatment modalities including immunotherapy [44]. A recent study revealed that NSCLC patients with KRAS mutations and concomitant STK11 and/or KEAP1 mutations had poorer prognoses. These patients may derive greater benefits from a combination of chemotherapy, atezolizumab, and bevacizumab (ABCP), as opposed to chemotherapy combined with atezolizumab/bevacizumab (ACP or BCP) [45]. However, the lack of available co-mutation information in this study is attributed to the fact that whole exome sequencing was only performed in a minority of patients. Therefore, further investigations focusing on co-mutations associated with driver oncogenes are still required.
Despite the advantages of our study, its retrospective nature has imposed certain limitations. Firstly, the sample size was relatively small and the follow-up period was not long enough. Nonetheless, given that the efficacy of neoadjuvant immunotherapy for resectable and locally advanced NSCLCs was still in its exploratory phase, our use of pathological response information allowed us to demonstrate the efficacy of immunotherapy more precisely and intuitively. Secondly, the exploratory of response of non-KRAS driver mutations to immunotherapy is still insufficient. Owing to the limitations of test kits and sample size, this study focused solely on the seven major oncogenic driver mutations. Other oncogenic driver mutations, including NTRK, NRG1, HRAS, NRAS, MEK1 need further study to provide a more comprehensive analysis of their response to immunotherapy. Thirdly, whole exome sequencing and detailed information regarding KRAS-mutant subtypes were only available in a minority of patients, which constrained our ability to conduct comprehensive investigations in these domains. Finally, less than half of the patients had pre-treatment PD-L1 expression results, limiting our research on the correlation between driver oncogene status and PD-L1 expression.
In summary, oncogenic driver mutations exert various impacts on the TIME of NSCLCs, thereby influencing the clinical benefit of ICIs. Our study found that patients with KRAS mutations were not only associated with a better pathological response to neoadjuvant immunotherapy in terms of oncogenic driver mutations but also correlated with better prognoses among resectable NSCLC patients. Increased tumor immunogenicity and a higher proportion of infiltrating CD8+ TILs were associated with KRAS driver mutations, which may explain the better response in immunotherapy. Consequently, molecular testing is necessary for patients scheduled to receive neoadjuvant treatment, and further prospective studies are still needed in the future.
Supplementary Information
Below is the link to the electronic supplementary material.
Author contributions
Concept and design were contributed by ZS, LZ and PZ; Acquisition of data was contributed by ZS, MT, DB, JZ, XZ, YQ, SH, YC, WY and HY; Analysis and interpretation of clinical data and RNA sequencing data were contributed by ZS, MT and LH; Drafting of the manuscript was contributed by ZS and LZ. Critical revision was contributed by ZS, MT, LH, LZ and PZ. Funding acquisition was contributed by ZS and PZ. All authors contributed to the article and approved the submitted version.
Funding
This research was supported by the National Natural Science Foundation of China (Grant no. 82125001), the Innovation Program of Shanghai Municipal Education Commission (Grant no. 2023ZKZD33), Clinical Research foundation of Shanghai Pulmonary Hospital (Grant no. FKLY20004), and Science and Technology Commission of Shanghai Municipality (Grant no. 23YF1435200).
Data availability
RNA sequencing data were deposited in the Genome Sequence Archive database under accession number HRA002071. Data and codes utilized in this study are immediately available from the corresponding author upon reasonable request.
Declarations
Conflict of interest
The authors have declared that no conflict of interest exists.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Ziyun Shen, Meixin Teng and Lu Han contributed equally to this work.
Contributor Information
Lele Zhang, Email: murongdule@163.com.
Peng Zhang, Email: zhangpeng1121@tongji.edu.cn.
References
- 1.Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48. doi: 10.3322/caac.21763. [DOI] [PubMed] [Google Scholar]
- 2.Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr, Wu YL, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299–311. doi: 10.1016/S0140-6736(16)30958-8. [DOI] [PubMed] [Google Scholar]
- 3.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. Nivolumab plus Ipilimumab in advanced non-small-cell lung cancer. N Engl J Med. 2019;381(21):2020–2031. doi: 10.1056/NEJMoa1910231. [DOI] [PubMed] [Google Scholar]
- 5.Blumenthal GM, Bunn PA, Jr, Chaft JE, McCoach CE, Perez EA, Scagliotti GV, et al. Current status and future perspectives on neoadjuvant therapy in lung cancer. J Thorac Oncol. 2018;13(12):1818–1831. doi: 10.1016/j.jtho.2018.09.017. [DOI] [PubMed] [Google Scholar]
- 6.Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med. 2022;386(21):1973–1985. doi: 10.1056/NEJMoa2202170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Leal TA, Ramalingam SS. Neoadjuvant therapy gains FDA approval in non-small cell lung cancer. Cell Rep Med. 2022;3(7):100691. doi: 10.1016/j.xcrm.2022.100691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.National Comprehensive Cancer Network. Non-Small Cell Lung Cancer (Version 2.2023). https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf.
- 9.Hanna NH, Robinson AG, Temin S, Baker S, Jr, Brahmer JR, Ellis PM, et al. Therapy for stage IV non-small-cell lung cancer with driver alterations: ASCO and OH (CCO) joint guideline update. J Clin Oncol. 2021;39(9):1040–1091. doi: 10.1200/JCO.20.03570. [DOI] [PubMed] [Google Scholar]
- 10.Zhao ZR, Lin ZC, Shen JF, Xie ZH, Jiang L. Neoadjuvant immunotherapy in oncogene-positive non-small cell lung cancer: a multicenter study. Ann of Thorac Surg. 2023;116(4):703–710. doi: 10.1016/j.athoracsur.2022.11.035. [DOI] [PubMed] [Google Scholar]
- 11.Chaft JE, Oezkan F, Kris MG, Bunn PA, Wistuba II, Kwiatkowski DJ, et al. Neoadjuvant atezolizumab for resectable non-small cell lung cancer: an open-label, single-arm phase II trial. Nat Med. 2022;28(10):2155–2161. doi: 10.1038/s41591-022-01962-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhang C, Chen HF, Yan S, Wu L, Yan LX, Yan XL, et al. Induction immune-checkpoint inhibitors for resectable oncogene-mutant NSCLC: a multicenter pooled analysis. NPJ Precis Oncol. 2022;6(1):66. doi: 10.1038/s41698-022-00301-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934–949. doi: 10.1016/j.cell.2017.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Otano I, Ucero AC, Zugazagoitia J, Paz-Ares L. At the crossroads of immunotherapy for oncogene-addicted subsets of NSCLC. Nat Rev Clin Oncol. 2023;20(3):143–159. doi: 10.1038/s41571-022-00718-x. [DOI] [PubMed] [Google Scholar]
- 15.Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol. 2019;30(8):1321–1328. doi: 10.1093/annonc/mdz167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lisberg A, Cummings A, Goldman JW, Bornazyan K, Reese N, Wang T, et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor Naive patients with advanced NSCLC. J Thorac Oncol. 2018;13(8):1138–1145. doi: 10.1016/j.jtho.2018.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang P, Dai J, Sun F, Xia H, He W, Duan L, et al. Neoadjuvant sintilimab and chemotherapy for resectable stage IIIA non-small cell lung cancer. Ann Thorac Surg. 2022;114(3):949–958. doi: 10.1016/j.athoracsur.2022.01.039. [DOI] [PubMed] [Google Scholar]
- 18.Zhu X, Sun L, Song N, He W, Xie B, Hu J, et al. Safety and effectiveness of neoadjuvant PD-1 inhibitor (toripalimab) plus chemotherapy in stage II-III NSCLC (LungMate 002): an open-label, single-arm, phase 2 trial. BMC Med. 2022;20(1):493. doi: 10.1186/s12916-022-02696-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–457. doi: 10.1038/nmeth.3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Yu Y, Zeng D, Ou Q, Liu S, Li A, Chen Y, et al. Association of survival and immune-related biomarkers with immunotherapy in patients with non-small cell lung cancer: a meta-analysis and individual patient-level analysis. JAMA Netw Open. 2019;2(7):e196879. doi: 10.1001/jamanetworkopen.2019.6879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Liu C, Zheng S, Jin R, Wang X, Wang F, Zang R, et al. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett. 2020;470:95–105. doi: 10.1016/j.canlet.2019.10.027. [DOI] [PubMed] [Google Scholar]
- 22.Anderson NR, Minutolo NG, Gill S, Klichinsky M. Macrophage-based approaches for cancer immunotherapy. Cancer Res. 2021;81(5):1201–1208. doi: 10.1158/0008-5472.CAN-20-2990. [DOI] [PubMed] [Google Scholar]
- 23.Hu J, Zhang L, Xia H, Yan Y, Zhu X, Sun F, et al. Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. 2023;15(1):14. doi: 10.1186/s13073-023-01164-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Qiu Y, Chen T, Hu R, Zhu R, Li C, Ruan Y, et al. Next frontier in tumor immunotherapy: macrophage-mediated immune evasion. Biomark Res. 2021;9(1):72. doi: 10.1186/s40364-021-00327-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.El Osta B, Behera M, Kim S, Berry LD, Sica G, Pillai RN, et al. Characteristics and outcomes of patients with metastatic KRAS-mutant lung adenocarcinomas: the lung cancer mutation consortium experience. J Thorac Oncol. 2019;14(5):876–889. doi: 10.1016/j.jtho.2019.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Liu SY, Sun H, Zhou JY, Jie GL, Xie Z, Shao Y, et al. Clinical characteristics and prognostic value of the KRAS G12C mutation in Chinese non-small cell lung cancer patients. Biomark Res. 2020;8:22. doi: 10.1186/s40364-020-00199-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Slebos RJ, Kibbelaar RE, Dalesio O, Kooistra A, Stam J, Meijer CJ, et al. K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung. N Engl J Med. 1990;323(9):561–565. doi: 10.1056/NEJM199008303230902. [DOI] [PubMed] [Google Scholar]
- 28.Passiglia F, Cappuzzo F, Alabiso O, Bettini AC, Bidoli P, Chiari R, et al. Efficacy of nivolumab in pre-treated non-small-cell lung cancer patients harbouring KRAS mutations. Br J Cancer. 2019;120(1):57–62. doi: 10.1038/s41416-018-0234-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jeanson A, Tomasini P, Souquet-Bressand M, Brandone N, Boucekine M, Grangeon M, et al. Efficacy of immune checkpoint inhibitors in KRAS-mutant non-small cell lung cancer (NSCLC) J Thorac Oncol. 2019;14(6):1095–1101. doi: 10.1016/j.jtho.2019.01.011. [DOI] [PubMed] [Google Scholar]
- 30.Liu Y, Zhang Z, Rinsurongkawong W, Gay CM, Le X, Ning MS, et al. Association of driver oncogene variations with outcomes in patients with locally advanced non-small cell lung cancer treated with chemoradiation and consolidative durvalumab. JAMA Netw Open. 2022;5(6):e2215589. doi: 10.1001/jamanetworkopen.2022.15589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Salgia R, Pharaon R, Mambetsariev I, Nam A, Sattler M. The improbable targeted therapy: KRAS as an emerging target in non-small cell lung cancer (NSCLC) Cell Rep Med. 2021;2(1):100186. doi: 10.1016/j.xcrm.2020.100186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wang Y, Li C, Wang Z, Wang Z, Wu R, Wu Y, et al. Comparison between immunotherapy efficacy in early non-small cell lung cancer and advanced non-small cell lung cancer: a systematic review. BMC Med. 2022;20(1):426. doi: 10.1186/s12916-022-02580-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Dong ZY, Zhong WZ, Zhang XC, Su J, Xie Z, Liu SY, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2017;23(12):3012–3024. doi: 10.1158/1078-0432.CCR-16-2554. [DOI] [PubMed] [Google Scholar]
- 34.Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell. 2016;165(7):1818. doi: 10.1016/j.cell.2016.05.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRAS(G12C) inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020;383(13):1207–1217. doi: 10.1056/NEJMoa1917239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Qiao M, Jiang T, Liu X, Mao S, Zhou F, Li X, et al. Immune checkpoint inhibitors in EGFR-mutated NSCLC: Dusk or dawn? J Thorac Oncol. 2021;16(8):1267–1288. doi: 10.1016/j.jtho.2021.04.003. [DOI] [PubMed] [Google Scholar]
- 37.Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z, et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res. 2016;22(18):4585–4593. doi: 10.1158/1078-0432.CCR-15-3101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lv C, Fang W, Wu N, Jiao W, Xu S, Ma H, et al. Osimertinib as neoadjuvant therapy in patients with EGFR-mutant resectable stage II-IIIB lung adenocarcinoma (NEOS): a multicenter, single-arm, open-label phase 2b trial. Lung Cancer. 2023;178:151–156. doi: 10.1016/j.lungcan.2023.02.011. [DOI] [PubMed] [Google Scholar]
- 39.Zhong WZ, Yan HH, Chen KN, Chen C, Gu CD, Wang J, et al. Erlotinib versus gemcitabine plus cisplatin as neoadjuvant treatment of stage IIIA-N2 EGFR-mutant non-small-cell lung cancer: final overall survival analysis of the EMERGING-CTONG 1103 randomised phase II trial. Signal Transduct Target Ther. 2023;8(1):76. doi: 10.1038/s41392-022-01286-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Negrao MV, Skoulidis F, Montesion M, Schulze K, Bara I, Shen V, Xu H, Hu S, Sui D, Elamin YY, Le X. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J Immunother Cancer. 2021;9(8):e002891. doi: 10.1136/jitc-2021-002891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med. 2014;371(23):2167–2177. doi: 10.1056/NEJMoa1408440. [DOI] [PubMed] [Google Scholar]
- 42.Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med. 2017;377(9):829–838. doi: 10.1056/NEJMoa1704795. [DOI] [PubMed] [Google Scholar]
- 43.Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer. 2019;19(9):495–509. doi: 10.1038/s41568-019-0179-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 2015;5(8):860–877. doi: 10.1158/2159-8290.CD-14-1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.West HJ, McCleland M, Cappuzzo F, Reck M, Mok TS, Jotte RM, et al. Clinical efficacy of atezolizumab plus bevacizumab and chemotherapy in KRAS-mutated non-small cell lung cancer with STK11, KEAP1, or TP53 comutations: subgroup results from the phase III IMpower150 trial. J Immunother Cancer. 2022;10(2):e003027. doi: 10.1136/jitc-2021-003027. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
RNA sequencing data were deposited in the Genome Sequence Archive database under accession number HRA002071. Data and codes utilized in this study are immediately available from the corresponding author upon reasonable request.