Your new experience awaits. Try the new design now and help us make it even better

ORIGINAL RESEARCH article

Front. Oncol., 27 November 2025

Sec. Thoracic Oncology

Volume 15 - 2025 | https://doi.org/10.3389/fonc.2025.1675825

This article is part of the Research TopicReal-World Data and Real-World Evidence in Lung Cancer Volume IIView all 18 articles

Impact of KRAS G12C mutation on the efficacy of chemoradiotherapy in patients with unresectable stage II or III non-small cell lung cancer

Jean Cabon*Jean Cabon1*Delphine LerougeDelphine Lerouge2Sebastien ThureauSebastien Thureau3Jacques BalossoJacques Balosso2Marin GuigoMarin Guigo2Radj GervaisRadj Gervais1Catherine DubosCatherine Dubos1Pascal DPascal Dô1Pierre DemontrondPierre Demontrond1Hubert CurcioHubert Curcio1Simon DeshayesSimon Deshayes4Jeannick MadelaineJeannick Madelaine4Dimitri Leite FerreiraDimitri Leite Ferreira4Leonard JacsonLeonard Jacson4Kilian LecrosnierKilian Lecrosnier5Franois ChevalierFrançois Chevalier5Florian GuisierFlorian Guisier6Edouard DantoingEdouard Dantoing6Alexandra LeconteAlexandra Leconte7Franois ChristyFrançois Christy7Mathieu Csaire,Mathieu Césaire2,5
  • 1Department of Medical Oncology, Centre François Baclesse, Caen, France
  • 2Department of Radiation Oncology, Centre François Baclesse, Caen, France
  • 3Department of Radiation Oncology, Centre Henri Becquerel, Rouen, France
  • 4Department of Pneumology, Centre Hospitalier Universitaire de Caen, Caen, France
  • 5UMR6252 CIMAP (Research Center for Ions, Materials, and Photonics), Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, Caen, France
  • 6Department of Pneumology, Centre Hospitalier Universitaire de Rouen, Rouen, France
  • 7Department of Clinical Research, Centre François Baclesse, Caen, France

Background: Approximately 35% of patients with non-small cell lung cancer (NSCLC) have locally advanced disease. Despite treatment with chemoradiotherapy (CRT) and consolidation immunotherapy, overall survival remains below 50% at 5 years. Kirsten rat sarcoma (KRAS) mutations (KRASms) are the most common lung cancer mutations, affecting 25% of NSCLC cases. KRASm can cause radioresistance, and several targeted KRASm therapies have been developed, mainly targeting the KRAS G12C mutation (KRASm G12C), but the impact of KRASm G12C on the efficacy of CRT for locally advanced NSCLC remains unclear.

Methods: We conducted a multicenter retrospective study of unresectable stage II or III NSCLC treated with CRT in four French hospitals between January 2014 and December 2022. The primary endpoint was the objective response rate (ORR) for KRASm G12C compared to KRAS wild-type (KRASwt). The main secondary objectives were to assess the difference in ORR between KRASm and KRASwt, and the difference in disease control rate (DCR), overall survival (OS), progression-free survival (PFS), time to local relapse (TTLR), and time to distant relapse (TTDR) according to KRASm status.

Results: Our study included 267 patients, and 73 patients had KRASm (27.3%). The most common KRASm was KRASm G12C (n = 42). Tumors were lung adenocarcinoma in 91% (n = 244) of patients. Two hundred (75%) patients were treated with concomitant CRT. There was no difference between KRASm G12C and KRASwt patients in terms of ORR (48% vs. 49%; p = 0.961) and DCR (86% vs. 84%; p = 0.903), nor when comparing KRASm to KRASwt in terms of OS (p = 0.64), PFS (p = 0.28), TTLR (p = 0.26), and TTDR (p = 0.3), with no impact after adjustment for durvalumab.

Conclusion: KRAS G12C mutation compared to KRAS wild-type did not affect response to chemoradiotherapy, and KRAS mutations compared to KRAS wild-type were not associated with worse survival in unresectable stage II or III NSCLC treated with chemoradiotherapy.

Highlights

KRASms are the most common lung cancer oncogene mutations in NSCLC (25% of NSCLC cases), with a predominance of KRASm G12C in approximately 40%.

KRAS G12C mutation, compared to KRAS wild-type, did not affect response to CRT.

KRAS mutations compared to KRAS wild-type were not associated with worse survival in unresectable stage II or III NSCLC treated with CRT.

Introduction

Lung cancer is a real public health problem worldwide. It is estimated to be the leading cause of cancer-related death, with 1.8 million deaths by 2020 (1). Non-small cell lung cancer (NSCLC) accounts for more than 80% of lung cancer cases, and approximately 35% of patients with NSCLC have locally advanced disease. Despite treatment with chemoradiotherapy (CRT) and consolidation immunotherapy, overall survival remains below 50% at 5 years in patients with unresectable stage III NSCLC (2). Lung cancer is a molecularly heterogeneous disease, and understanding its biology is crucial for the development of effective therapies (3).

Kirsten rat sarcoma (KRAS) mutations (KRASms) are the most frequent lung cancer oncogenic mutations (25% of NSCLC and 30%–37% of adenocarcinoma cases), with the majority of cases involving codons 12 and 13 (4). Ras proteins are encoded by three ubiquitously expressed genes: HRAS, KRAS, and NRAS. KRAS is the most frequently mutated, followed by NRAS (5). These proteins are GTPases that function as molecular switches regulating pathways, responsible for cell proliferation and survival. Aberrant Ras function is associated with hyperproliferative developmental disorders and cancer (6). Among KRASms in NSCLC, the KRAS G12C mutation (KRASm G12C) is the most common (approximately 40%), followed by the KRAS G12V mutation (20%) and the KRAS G12D mutation (16%) (7, 8). KRASms are associated with smoking history and Caucasian ethnicity, with a higher proportion of never smokers for KRAS G12D compared to KRASm G12C and KRAS G12V mutations (9, 10).

Several studies have reported a negative impact of KRASm G12C on survival, but the prognostic impact remains controversial, with conflicting data in the literature (1114).

In addition, KRASm may cause radioresistance, although the exact mechanisms remain unknown (1518). In a retrospective study of stage III NSCLC treated with CRT, KRASms were associated with a lower response rate (63% vs. 81%), which may indicate a reduced efficacy of CRT (19). However, the study included 119 patients with only 13% KRASms and did not specify the subtype of KRASm.

Several targeted therapies have been developed, mainly targeting the KRASm G12C, such as sotorasib or adagrasib, which currently have a therapeutic impact in clinical practice for metastatic lung cancer (20, 21). Other therapies targeting the KRASm G12C or other KRAS mutations, as single agents or in combination, are being investigated in clinical trials (22).

Because of the hypothetical radioresistance of KRASm that may reduce the efficacy of CRT and the existence of targeted KRASm G12C therapies that may counteract these resistance mechanisms, our study aimed to evaluate the impact of KRASm on the efficacy of CRT in patients with unresectable stage II or III NSCLC.

Methods

Objectives

The primary objective was to evaluate the response rate to CRT in patients with unresectable stage II or III NSCLC according to KRASm G12C status. The primary endpoint was the objective response rate (ORR), assessed using the number of complete response (CR) or partial response (PR) compared to the total number of patients for KRASm G12C NSCLC compared to KRAS wild-type (KRASwt). The ORR was chosen as the best way to evaluate response to CRT, particularly in relation to the hypothetical radioresistance induced by KRASm. ORR at the first assessment was not affected by the addition of durvalumab, unlike survival data. The secondary objectives were to assess the differences in ORR and disease control rate (DCR) to chemoradiotherapy for KRASm compared to KRAS wild-type, KRASm G12C compared to KRASm non-G12C, and KRASm compared to KRASwt. Another aim was to evaluate the impact of KRASm on overall survival (OS), progression-free survival (PFS), time to local relapse (TTLR), and time to distant relapse (TTDR). Finally, the impact of main co-mutations, such as KRAS/TP53 (KP) and KRAS/STK11 (KL), on the response to CRT, as well as their impact on OS and PFS, was evaluated.

Patient selection

A retrospective observational multicenter study of unresectable stage II or III NSCLC treated with CRT was conducted in four French hospitals between January 2014 and December 2022. The inclusion criteria were age ≥ 18 years, patients with unresectable stage II or III NSCLC and treated with concomitant or sequential CRT, and patients who did not object to the use of their medical data for cancer research purposes. The exclusion criteria were the absence of KRASm status (molecular biology not performed or not available), patient refusal to participate, the absence of chemotherapy, and total radiotherapy dose <60 Gy isoeffective in 2-Gy fractions (EQD2), as this threshold corresponds to a curative dose in the included patients. Genomic testing for KRASm was performed using next-generation sequencing (NGS) or broad panels based on polymerase chain reaction. This study was conducted in accordance with the French Research Standard MR-004 “Research not involving human participants” and is registered with the French Health Data Hub under the reference F20211126103028.

Data collection

Patient characteristics included the World Health Organization (WHO) performance status prior to CRT, smoking habit (never, current, and former), and demographic data such as age and sex. Former smokers were defined as those who ceased smoking over a period of at least 1 year. Lung cancer characteristics included histology, TNM stage, positron emission tomography (PET) scan imaging, history of lung cancer, history of lung radiotherapy, history of lung cancer surgery, and programmed death-ligand 1 (PD-L1) status expressed as a percentage of positive tumor cells: negative (<1%), low (1%–50%), and high (>50%). Collected radiotherapy modalities were treatment duration, total dose, dose per fraction, number of fractions, and technique (3D conformal versus intensity-modulated radiotherapy). Data on chemotherapy included concomitant or sequential chemotherapy, the type of chemotherapy, the presence of induction chemotherapy only for patients treated with a concomitant strategy, and the number of cycles of platinum-based chemotherapy (one cycle of platinum-based chemotherapy according to the following charts: D1 = D22 or D1, D8, D15, D1 = D22). The following were also collected: mutation status, presence of consolidation immunotherapy, date of relapse (regardless of local or metastatic relapse site), date of local relapse, date of metastatic relapse, date of death, and causes of death. Treatment response was assessed using CT scan reports of complete or partial response, and stable or relapsed disease, based on Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. The response to treatment was evaluated based on the first CT scan performed after CRT.

Statistical analysis

ORR was defined as the percentage of patients who achieved a response, which could be either a complete response or a partial response. DCR was defined as the percentage of patients who achieved a response or stability. PFS was defined from the start of chemotherapy until the date of relapse, date of death of any cause, or date of last news. OS was defined from the start of chemotherapy until death from any cause or last news. TTLR was defined as the time from the start of chemotherapy to local relapse, and TTDR was defined as the time from the start of chemotherapy to distant relapse.

Descriptive statistics were mean, standard deviation, median, extreme values for continuous data, and frequencies and percentages for categorical data.

OS, PFS, TTLR, and TTDR were estimated using the Kaplan–Meier method, and comparisons were made using the log-rank test. Proportional Cox regression was used to evaluate the association between time-to-event outcomes and covariates.

All tests were two-sided; p-values <0.05 were considered statistically significant. All analyses were performed in R version 4.1.2.

Results

Description of the study population

A total of 267 patients with unresectable stage II or III NSCLC treated with CRT were included in our study (Figure 1), of which 73 had KRASm (27.3%). The three most common KRASms were KRAS G12C (n = 42, 15.7%), KRAS G12V (n = 13, 4.9%), and KRAS G12D (n = 7, 2.6%). In the KRASm population, a co-mutation with TP53 and STK11 in cases where the mutation status was known was reported in 45.5% (n = 20/44) and 7% (n = 4/57), respectively. For molecular alterations for which molecular status was available, excluding KRASm, the most commonly reported molecular alterations (with a prevalence >5%) were TP53 in 58.1% (n = 97/167), BRAF in 6.4% (n = 17/265), STK11 in 6.4% (n = 14/219), and EGFR in 5.6% (n = 15/266) (Supplementary Table 1, Supplementary Figure 1). The median age was 63.8 years (range, 30.9–83.0), and there was a male predominance (n = 177, 66%). Performance status (PS) was 0 or 1 for 96% (n = 257) and a current or former smoking status for 94% (n = 244) of patients. Lung adenocarcinoma was observed in 91% (n = 244), and the remainder were not otherwise specified (NOS) NSCLC (n = 23, 9%). Eight percent (n = 21) of patients had stage II disease (n = 21) and 92% (n = 232) stage III, with a predominance of stage IIIA (n = 101, 40%) and stage IIIB (n = 104, 41%). Seventy-five percent (n = 200) of patients were treated with concomitant CRT, and induction chemotherapy was given in 90% (n = 180) of the patients. All but one patient received platinum-based concurrent chemotherapy, predominantly carboplatin only (n = 210, 78.5%), with a mean number of platinum cycles of 3.67 (range, 0–8 cycles). Taxane chemotherapy was reported in 54% (n = 144) and pemetrexed in 49% (n = 130) (Supplementary Table 2). The median duration of radiotherapy was 47 days (range, 32–92), and the median dose was 66 Gy (range, 55–86) with a median dose per fraction of 2 Gy (range, 2–2.75). PD-L1 status was negative, 1%–49%, >50%, and unknown in 40% (n = 74), 35% (n = 64), 25% (n = 45), and 31.5% (n = 84), respectively. The administration of durvalumab consolidation was reported in 28% (n = 76) (Table 1 for general features of the population study).

Figure 1
Flowchart illustrating patient selection for a study on lung cancer treated with thoracic radiotherapy from January 2014 to December 2022. Initially, 1,146 patients were eligible; 188 were excluded due to small cell lung cancer (SCLC). Of 958 non-small-cell lung cancer (NSCLC) patients, further exclusions included lack of mKRAS status, no chemotherapy, insufficient radiotherapy dose, metastatic status, no histology, missing CRT response data, or duplicate cases. Ultimately, 267 patients with unresectable stage II or III NSCLC and treated with chemoradiotherapy (CRT) were included.

Figure 1. Study flowchart. SCLC, small cell lung cancer; NSCLC, non-small cell lung cancer; KRASm, KRAS mutation; CRT, chemoradiotherapy.

Table 1
www.frontiersin.org

Table 1. General features of the study population.

KRASm and therapeutic response to chemoradiotherapy (KRASm G12C, KRASm non-G12C, and KRASm)

There was no difference between KRASm G12C and KRASwt patients in terms of ORR (48% vs. 49%; p = 0.961) and DCR (86% vs. 84%; p = 0.903) (Supplementary Table 3).

There was no difference between KRASm and KRASwt patients in terms of ORR (47% vs. 49%; p = 0.78) and DCR (88% vs. 84%; p = 0.515) (Supplementary Table 4).

There was no difference between KRASm G12C and KRASm non-G12C in terms of ORR (48% vs. 45%); p = 1) and DCR (86% vs. 90%; p = 0.724) (Supplementary Table 5).

No difference was observed in the ORR or DCR between KRASwt, KRASm G12C, and KRASm non-G12C (p = 0.895 for ORR and p = 0.664 for DCR) (Table 1).

KRASm and survival

There was no difference in OS between KRASm and KRASwt (p = 0.64). However, there was a numerical benefit for KRASm with a median OS (mOS) of 62.7 months for KRASm (95% CI, 28.3–NA) and an estimated 5-year survival rate of 54.3%. In contrast, the mOS was 47.2 months (95% CI, 34.2–64.4), and the estimated 5-year survival rate was 41.2% for KRASwt patients (Figure 2). On multivariate analysis, there was no association between KRASm and OS [hazard ratio (HR) = 1.13, p = 0.61] (Figure 3).

Figure 2
Kaplan-Meier survival curve showing overall survival probability over time in months for KRAS wild-type (blue line) and KRAS mutation (red line). The p-value is 0.64, indicating no significant difference. Below the curve, numbers at risk are provided at different time points for both KRAS groups.

Figure 2. OS according to KRAS mutation status in NSCLC treated with chemoradiotherapy. OS, overall survival; KRASwt, KRAS wild-type; KRASm, KRAS mutation; NSCLC, non-small cell lung cancer. p-Value considered statistically significant was less than 0.05.

Figure 3
Forest plot illustrating hazard ratios for overall survival based on various factors, including age, WHO status, cancer stage, chemotherapy type, and treatments. Significant values include WHO status 1 and 2, Stage IIIB, Stage IIIC, number of platinum salt cycles, and Durvalumab, with p-values below 0.05, indicating statistical significance.

Figure 3. Prognostic factors associated with OS in multivariate analysis. OS, overall survival; HR, hazard ratio; KRASm, KRAS mutation; KRASwt, KRAS wild-type. p-Value considered statistically significant was less than 0.05.

There was no difference in PFS between KRASm and KRASwt (p = 0.28), with median PFS of 13.8 months for KRASm (95% CI, 10.8–29.6) and 12.7 months for KRASwt (95% CI, 11.0–15.2), and estimated 5-year PFS of 27% for KRASm and 19.5% for KRASwt (Figure 4). On multivariate analysis, there was no association between KRASm and PFS (HR = 1.01, p > 0.9) (Figure 5).

Figure 4
Kaplan-Meier survival curve showing progression-free survival probability over time in months for KRAS wild-type (blue) and KRAS mutation (red) groups. The p-value is 0.28, indicating no statistical significance. The numbers at risk are shown below the time axis.

Figure 4. PFS according to KRAS mutation status in NSCLC treated with chemoradiotherapy. PFS, progression-free survival; KRASwt, KRAS wild-type; KRASm, KRAS mutation; NSCLC, non-small cell lung cancer. p-Value considered statistically significant was less than 0.05.

Figure 5
A forest plot showing hazard ratios (HR) and confidence intervals (CI) for progression-free survival (PFS) related to various factors. Notable significant factors include Stage IIIB and IIIC, PD-L1 ≤50%, Durvalumab, and STK11 mutation, with p-values less than 0.05.

Figure 5. Prognostic factors associated with PFS in multivariate analysis. PFS, progression-free survival; WHO status, World Health Organization status; PD-L1, programmed death-ligand 1; KRASm, KRAS mutation; STK11m, STK11 mutation; HR, hazard ratio. p-Value considered statistically significant was less than 0.05.

No difference was observed in OS or PFS between KRASwt, KRASm G12C, and KRASm non-G12C (p = 0.89 for OS and p = 0.27 for PFS) (Supplementary Figures 2, 3).

KRASm and local/distant relapses

There was no difference in TTLR when comparing KRASm to KRASwt (p = 0.26), with a median TTLR of 26.9 months (95% CI, 18.3–40.3) for KRASm and 18.2 months for KRASwt (95% CI, 15.2–26.5) (Supplementary Figure 4). There was no difference in TTDR (p = 0.3) with a median of 25.9 months (95% CI, 16.4–NA) for KRASm and 17.1 months for KRASwt (95% CI, 13.8–26.5) (Supplementary Figure 5).

On multivariate analysis, there was no association between KRASm and TTLR (HR = 0.86, p = 0.4) or TTDR (HR = 0.86, p = 0.44) (Supplementary Figure 6 for TTLR and Supplementary Figure 7 for TTDR).

Results for durvalumab (subgroup analysis and all cases)

On multivariate analysis, durvalumab was associated with better OS (HR = 0.21, p < 0.001), PFS (HR = 0.3, p < 0.001), TTLR (HR = 0.39, p < 0.001), and TTDR (HR = 0.32, p < 0.001) (Figures 3, 5; Supplementary Figures 6, 7).

There was no difference in OS, PFS, TTLR, and TTDR after adjusting for the presence of durvalumab when comparing KRASm to KRASwt (OS, p = 0.44; PFS, p = 0.97; TTLR, p =0.65; TTDR, p = 0.86) (Supplementary Figures 8a, 9a, 10a, 11a).

For all patients treated with durvalumab, durvalumab was associated with better OS (p < 0.0001) and PFS (p < 0.0001) compared to patients not treated with durvalumab, with estimated 5-year rates of 75.3% and 37.7% for OS and PFS, respectively (Supplementary Figures 12, 13).

The impact of KP and KL on therapeutic response and survival outcomes (OS and PFS)

There were no differences observed between KP and KP wild-type (KPwt) in terms of ORR (45% vs. 50%; p = 0.92), but there was a significant trend in favor of the KP group regarding DCR (100% vs. 85%; p = 0.09) (Supplementary Figure 14). There was also no significant difference in OS and PFS, although there was a trend toward better PFS in favor of the KP group (p = 0.64 for OS and 0.12 for PFS) (Supplementary Figures 15, 16 for OS and PFS, respectively).

There were no differences in therapeutic response or OS between KL and KL wild-type (KLwt) (ORR, p = 0.62; DCR, p = 1; OS, p = 0.84). However, PFS was worse in the KL group (p = 0.029) (Supplementary Figures 1719 for therapeutic response, OS, and PFS, respectively).

Prognostic factors associated with OS and PFS

The results of the univariate analysis are available in Supplementary Figure 20.

As previously mentioned, on multivariate analysis, the presence of durvalumab in consolidation was associated with improved OS (HR = 0.21, p < 0.001) and a greater number of platinum salt cycles (HR = 0.73, p < 0.001). A poorer OS was observed in patients with more advanced disease (stage IIIB or IIIC) (IIIB, p = 0.04; IIIC, p = 0.009) and an impaired general condition (PS 1 or 2 vs. PS 0) (PS 1, p = 0.01; PS 2, p = 0.02).

The only factor associated with improved PFS was the presence of durvalumab (HR = 0.3, p < 0.001). The following factors were associated with poorer PFS: the presence of an STK11 mutation (HR = 4.81, p < 0.001), low PD-L1 status (HR = 2.29, p = 0.005), and advanced-stage IIIB/IIIC disease (IIIB, p = 0.03; IIIC, p =0.03).

Discussion

In our cohort of 267 patients with unresectable stage II or III NSCLC treated with CRT, approximately 27% had KRASm (n = 73), with a predominance of the KRASm G12C subtype (57.5% of all KRASm). These frequencies are consistent with those reported previously in large-scale molecular profiling studies of NSCLC (7, 8).No significant differences were observed between KRASm and KRASwt patients with respect to ORR (p = 0.78) or DCR (p = 0.515). Similarly, no differences were observed in terms of OS (p = 0.64), PFS (p = 0.28), TTLR (p = 0.26), and TTDR (p = 0.3) between KRASm and KRASwt. The prognostic impact of KRASm in NSCLC is still being debated. A meta-analysis of 41 studies involving 6,939 patients with NSCLC reported an unfavorable effect of KRASm on survival (HR = 1.45; 95% CI, 1.58-2.44). Subgroup analysis by stage revealed that KRASm was associated with poorer prognosis in early-stage disease: stage I (1.81; 95% CI, 1.36–2.39) and stage I–IIIa (1.68; 95% CI, 1.11–2.55), but not in advanced-stage disease (IIIb–IV) (1.3; 95% CI, 0.99–1.71). These findings suggest that KRAS mutations increase the risk of recurrence after resection but do not significantly affect the response to chemoradiotherapy and systemic therapies in stages III/IV (23). A retrospective study of 119 patients, including 16 with KRASm (13%), reported lower response rates to CRT among patients with KRASm compared to those with KRASwt (63% vs. 81%) (19). Another retrospective study of unresectable stage III NSCLC treated with CRT, including 114 KRASwt patients and 42 KRASm patients with 48% KRASm G12C (n = 20), reported that KRASm patients were less likely to receive immune checkpoint inhibitor (ICI) consolidation due to rapid disease progression post-CRT (23.8% vs. 4.4%, p = 0.007). In the Barsouk study, KRASm patients had worse PFS (median 6.3 vs. 10.7 months, p = 0.041) but similar OS (median 23.1 vs. 27.3 months, p = 0.237) (24).

However, the baseline characteristics of the studies varied. The Yagishita study included only Asian patients and had a male predominance (n = 12, 75%), whereas our cohort was balanced (58% of male patients), and the Barsouk study reported a predominance of female patients (n = 29, 69%).

In contrast, our study of a large cohort did not show any negative impact of KRASm on CRT response. Notably, prior studies did not specifically evaluate the impact of KRASm G12C on therapeutic outcomes. To our knowledge, our analysis is one of the few that directly evaluates this association. There were no significant differences in ORR (p = 0.961) or DCR (p = 0.903) between KRASm G12C and KRASwt patients. Likewise, OS and PFS did not differ among KRASwt, KRASm G12C, and KRASm non-G12C patients (p = 0.89 for OS and p = 0.27 for PFS). A recent systematic review and meta-analysis in NSCLC, KRASm G12C tumors had worse OS (HR = 1.42; 95% CI, 1.10–1.84, p = 0.007) but similar DFS (HR = 2.36, 95% CI 0.64–8.16) compared to KRASwt tumors. When compared to other KRAS mutations, KRASm G12C tumors had worse DFS (HR = 1.49; 95% CI, 1.07–2.09, p < 0.0001) but similar OS (HR = 1.03; 95% CI, 0.84–1.26). However, substantial heterogeneity and potential publication bias limit the robustness of these findings (11). Sebastian et al. analyzed a large real-world German cohort of 1,039 patients, including 160 patients with KRASm G12C (15.4%) and 251 patients with KRASm non-G12C (24.2%). They found no significant differences in clinical outcome between KRASwt, G12C, and non-G12C mutations; KRAS mutation status was not prognostic in the model. However, only a small proportion of NSCLC had non-metastatic disease (eight for KRASm G12C, 14 for KRASm non-G12C, and 43 for KRASwt). These findings are consistent with those of several other studies (2527).

We did not select either OS or PFS as the primary endpoint because the results would have been biased by the introduction of consolidation immunotherapy in 2017. In the PACIFIC study, durvalumab immunotherapy induced significant improvements in OS and PFS compared to placebo. The estimated 5-year OS and PFS rates were 42.9% and 33.1%, respectively, in the durvalumab arm (2). In our real-world study, durvalumab was associated with improved OS (HR = 0.21, p < 0.001) and PFS (HR = 0.3, p < 0.001). We observed a 5-year OS of 75.3% and a 5-year PFS of 37.7%. The comparable 5-year PFS and improved 5-year OS were attributed to the smaller sample size. After adjusting for durvalumab, we found no difference in OS (p = 0.44), PFS (p = 0.97), TTLR (p = 0.65), or TTDR (p = 0.86) between KRASm and KRASwt patients, with 26 KRASm patients receiving ICI consolidation therapy. Similar results were reported in the Barsouk study, which included 29 KRASm patients (PFS, 8.1 vs. 11.9 months, p = 0.35; OS, 30.5 vs. 31.7 months, p = 0.692), as well as in the Guo study, which included 18 KRASm patients (PFS, 12.6 vs. 12.7 months, p = 0.77; OS, 20.0 vs. 32.4 months, p = 0.69). However, divergent results were reported in the Liu study, which included 22 KRASm patients. These patients had worse PFS (8 vs. 40.1 months, p < 0.001) but similar OS (36.2 months vs. NA, p = 0.07) in the KRAS subgroup (24, 28, 29).

We will analyze the impact of the main co-mutations (KP and KL) in parallel. No differences were found in terms of DCR or ORR between KP and KPwt, nor between KL and KLwt (p = 0.09 and p = 1, respectively, for DCR; p = 0.92 and p = 0.84, respectively, for ORR). There were no differences in OS between KP and KPwt (p = 0.64), nor between KL and KLwt (p = 0.84). There was a trend toward better PFS in favor of the KP group (p = 0.12) and worse PFS in the KL group (p = 0.029). Additionally, we know that co-occurring genetic events can impact the immune environment. KP was associated with the increased expression of PD-L1 and mutational burden, showing a remarkable clinical benefit for PD-1 inhibitors (30), while KL was significantly associated with PD-L1 negative in tumor mutational burden intermediate-high and represents a major driver of primary resistance to PD-1 blockade in KRAS mutant (8). STK11 mutation was associated with a poorer prognosis (31), a finding that was also observed in our study. In the multivariate analysis, the STK11 mutation was associated with poorer PFS (HR = 4.81, p < 0.001). Poorer PFS was also found in the KL group, which may be explained by the lesser benefits from consolidation immunotherapy compared to KP switching. However, these data should be interpreted with caution, given the limited sample size of 20 KP patients and four KL patients in our study. Future studies with larger sample sizes are needed to confirm these findings.

Two KRASm G12C-targeting treatments have demonstrated efficacy and are currently in clinical use. The first, sotorasib, is a KRAS G12C inhibitor that significantly increased PFS and had a more favorable safety profile than docetaxel in patients with advanced NSCLC harboring the KRASm G12C mutation who had previously been treated with other anticancer drugs (20). The second is adagrasib, another KRAS G12C inhibitor. In a phase 2 study, it showed clinical activity with an objective response rate of 42.9%, a median duration of response of 8.5 months, and a median PFS of 6.5 months (21). Additionally, KRASm G12C tumors have been reported to exhibit higher PD-L1 expression than KRASwt tumors and may benefit from anti-PD-1/PD-L1 blockade (7, 3234). Several studies have reported promising results from combining a KRAS G12C inhibitor with an ICI (35, 36). Recently, the primary analysis of the KRYSTAL-12 trial, with a median follow-up of 9.4 months, showed a significant improvement in PFS with adagrasib versus docetaxel (HR = 0.58; 95% CI, 0.45–0.76; p < 0.0001; median PFS 5.49 vs. 3.84 months) in patients with KRASm G12C locally advanced or metastatic NSCLC who had previously received platinum-based chemotherapy, concurrently or sequentially with anti-PD-(L)1 therapy (37).

A recent phase III study showed that osimertinib treatment significantly increased progression-free survival (median PFS, 39.1 months for osimertinib vs. 5.6 months for placebo) in patients with unresectable stage III EGFR-mutated NSCLC who did not experience progression during or after chemoradiotherapy (38). A similar approach could be promising for a therapy targeting KRASm G12C in combination with immunotherapy. Several earlier-stage studies are currently evaluating a neoadjuvant strategy that combines a KRAS G12C inhibitor with either chemotherapy or immunotherapy (NCT05118854 and NCT05118854).

The present study has several limitations that are specific to retrospective studies. These limitations include the national scope of the study, the small size of the cohorts, and the real-world nature of the available molecular data, which varies between centers. Our study’s data do not suggest that KRASm G12C or KRASm is radioresistant. There is no significant difference in ORR compared to KRASwt. However, the results of our study may be influenced by limited statistical power resulting from the small patient sample size, and there may be other associated factors, such as co-mutations, that affect the response to CRT. PD-L1 status was unavailable for 31.5% of the patients in our study (n = 84), which can be explained by the fact that the therapeutic impact of PD-L1 was initially limited to patients with PD-L1 ≥ 1%. Furthermore, the impact of ICI consolidation on survival outcomes according to KRAS status was limited, as only 25 KRASm patients and 13 KRASm G12C patients were treated with durvalumab. All these data must be compared with prospective data from larger samples.

Conclusion

KRAS G12C mutation compared to KRAS wild-type did not affect response to chemoradiotherapy, and KRAS mutations compared to KRAS wild-type were not associated with worse survival in unresectable stage II or III NSCLC treated with chemoradiotherapy. Further studies are needed to investigate if co-mutations such as KP or KL could impact the response to chemoradiotherapy and survival in NSCLC.

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author.

Ethics statement

This study was conducted in accordance with the French Research Standard MR-004 “Research not involving Human participants” and is registered with the French Health Data Hub under the reference F20211126103028. The studies were conducted in accordance with the local legislation and institutional requirements. The participants provided their written informed consent to participate in this study. Written informed consent was obtained from the individual(s) for the publication of any potentially identifiable images or data included in this article.

Author contributions

JC: Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing. DL: Writing – review & editing. ST: Writing – review & editing. JB: Writing – review & editing. MG: Writing – review & editing. RG: Writing – review & editing. CD: Writing – review & editing. PDô: Writing – review & editing. PDe: Writing – review & editing. HC: Writing – review & editing. SD: Writing – review & editing. JM: Writing – review & editing. DF: Writing – review & editing. LJ: Writing – review & editing. KL: Writing – review & editing. FChe: Writing – review & editing. FG: Writing – review & editing. ED: Writing – review & editing. AL: Writing – review & editing. FChr: Formal analysis, Methodology, Software, Writing – review & editing. MC: Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2025.1675825/full#supplementary-material

Supplementary Table 1 | Mutation status prevalence KRASm, KRAS mutation; KP, KRAS/TP53 co-mutation; KL, KRAS/STK11 co-mutation; ALK, anaplastic lymphoma kinase; BRAF, v-Raf murine sarcoma viral oncogene homolog B; EGFR, epithelial growth factor receptor; HER2, human epidermal growth factor receptor 2; STK11, serine/threonine kinase 11; MET, mesenchymal epithelial transition; ROS-1, c-ROS oncogene 1; RET, rearranged during transfection; TP53, tumor protein 53; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic; there is a lack of data for certain genomic alterations due to heterogeneous molecular screening across centers for certain molecular alterations.

Supplementary Table 2 | concomitant chemotherapy with platinum salts and type of platinum salts. KRASm, mutated KRAS; KRASwt, KRAS wild-type; ICI, immune check-point inhibitor; VEGF, vascular endothelial growth factor; EGFR, epithelial Growth Factor Receptor; CRT, chemoradiotherapy; some patients may have been treated as part of a clinical trial.

Supplementary Table 3 | Comparison of KRASm G12C and KRASwt for response to chemoradiotherapy. KRASm G12c, KRAS G12C mutation; non-KRASm G12C, patients without KRAS G12C mutation; ORR, objective response rate; DCR, disease control rate; p-value considered statistically significant was less than 0.05.

Supplementary Table 4 | Comparison of KRASm and KRASwt for response to chemoradiotherapy. KRASm, KRAS mutation; KRASwt, wild-type KRAS; ORR, objective response rate; DCR, disease control rate; p-value considered statistically significant was less than 0.05.

Supplementary Table 5 | Comparison of KRASm G12C and KRASm non-G12C for response to chemoradiotherapy. KRASm G12c, KRAS G12C mutation; KRASm non-G12C, KRAS mutations excluding G12C; ORR, objective response rate; DCR, disease control rate; p-value considered statistically significant was less than 0.05.

Supplementary Figure 1 | Spectrum of KRASm. KRASm, KRAS mutation; other KRASm - KRASm other than those shown in figure. KRAS G12S, KRAS G12F and G13D were present in 1.4%.

Supplementary Figure 2 | OS comparison between KRASwt, KRASm G12C, and KRASm non-G12C. KRASm non-G12C, KRAS mutation, excluding KRAS G12C; KRASm G12C, KRAS G12C mutation KRASm G12C; KRASwt, KRAS wild-type; OS, overall survival; p-value considered statistically significant was less than 0.05.

Supplementary Figure 3 | PFS comparison between KRASwt, KRASm G12C, and KRASm non-G12C. KRASm non-G12C= KRAS mutation, excluding KRAS G12C; KRASm G12C, KRAS G12C mutation KRASm G12C; KRASwt, KRAS wild-type; PFS, progression-free survival; p-value considered statistically significant was less than 0.05.

Supplementary Figure 4 | TTLR according to KRAS mutation status in NSCLC treated by chemoradiotherapy. KRASwt, KRAS wild-type; KRASm, KRAS mutation; TTLR, time to local relapse; p-value considered statistically significant was less than 0.05.

Supplementary Figure 5 | TTDR according to KRAS mutation status in NSCLC treated by chemoradiotherapy. KRASwt, KRAS wild-type; KRASm, KRAS mutation; TTDR, time to distant relapse; p-value considered statistically significant was less than 0.05.

Supplementary Figure 6 | Prognostic factors associated with TTLR in multivariate analysis. TTLR, time to local relapse; WHO status, World Health Organization status; N platine, number of platinum salt cycles; HR, hazard ratio; CT, chemoradiotherapy; p-value considered statistically significant was less than 0.05.

Supplementary Figure 7 | Prognostic factors associated with TTDR in multivariate analysis. TTDR, time to distant relapse; WHO status, World Health Organization status; N platine, number of platinum salt cycles; HR, hazard ratio; IMRT, intensity-modulated radiation therapy; CT, chemoradiotherapy; KRASm, KRAS mutation; p-value considered statistically significant was less than 0.05.

Supplementary Figure 8 | Subgroup analysis for OS according to KRASm: durvalumab (a) and without durvalumab (b). (a). OS, overall survival; KRASwt, KRAS wild-type; KRASm= KRAS mutation; p-value considered statistically significant was less than 0.05. (b) OS, overall survival; KRASwt, KRAS wild-type; KRASm= KRAS mutation; p-value considered statistically significant was less than 0.05.

Supplementary Figure 9 | Subgroup analysis for PFS according to KRASm: durvalumab (a) and without durvalumab (b). (a) PFS, progression-free survival; KRASwt, KRAS wild-type; KRASm= KRAS mutation; p-value considered statistically significant was less than 0.05. (b) PFS, progression-free survival KRASwt, KRAS wild-type; KRASm= KRAS mutation; p-value considered statistically significant was less than 0.05.

Supplementary Figure 10 | Subgroup analysis for TTLR according to KRASm: durvalumab (a) and without durvalumab (b). (a) KRASwt, KRAS wild-type; KRASm= KRAS mutation; TTLR, time to local relapse; p-value considered statistically significant was less than 0.05. (b) KRASwt, KRAS wild-type; KRASm, KRAS mutation; TTLR, time to local relapse; p-value considered statistically significant was less than 0.05.

Supplementary Figure 11 | Subgroup analysis for TTDR according to KRASm: durvalumab (a) and without durvalumab (b). (a) KRASwt, KRAS wild-type; KRASm= KRAS mutation; TTDR= time to distant relapse; p-value considered statistically significant was less than 0.05. (b) KRASwt, KRAS wild-type; KRASm, KRAS mutation; TTDR, time to distant relapse; p-value considered statistically significant was less than 0.05.

Supplementary Figure 12 | Subgroup analysis for OS according to durvalumab treatment OS, overall survival; p-value considered statistically significant was less than 0.05.

Supplementary Figure 13 | Subgroup analysis for PFS according to durvalumab treatment. PFS, progression-free survival; p-value considered statistically significant was less than 0.05.

Supplementary Table 14 | Comparison of KP and KPwt for response to chemoradiotherapy. KP, KRAS/TP53 comutation; KPwt, Patients without KRAS or TP53 mutations; ORR, objective response rate; DCR, disease control rate; p-value considered statistically significant was less than 0.05.

Supplementary Table 15 | OS comparison between KP and KPwt. KP, KRAS/TP53 comutation; KPwt, Patients without KRAS or TP53 mutations; KPwt, both wild-type; WT, wild-type; OS, overall survival; p-value considered statistically significant was less than 0.05.

Supplementary Table 16 | PFS comparison between KP and KPwt. KP, KRAS/TP53 comutation; KPwt, Patients without KRAS or TP53 mutations; KPwt, both wild-type; WT, wild-type; PFS, progression-free survival; p-value considered statistically significant was less than 0.05.

Supplementary Table 17 | Comparison of KL and KLwt for response to chemoradiotherapy. KL, KRAS/STK11 comutation; KLwt, Patients without KRAS or STK11 mutations.; ORR, objective response rate; DCR, disease control rate; p-value considered statistically significant was less than 0.05.

Supplementary Table 18 | OS comparison between KL and KLwt. KL, KRAS/STK11 comutation; KLwt, Patients without KRAS or STK11 mutations; KLwt, both wild-type; WT, wild-type; OS, overall survival; p-value considered statistically significant was less than 0.05.

Supplementary Table 19 | PFS comparison between KL and KLwt. KL, KRAS/STK11 comutation; KLwt, Patients without KRAS or STK11 mutations; KLwt, both wild-type; WT, wild-type; PFS, progression-free survival; p-value considered statistically significant was less than 0.05.

Supplementary Table 20 | - results of the univariate analysis on OS, PFS, TTLR and TTDR. OS, overall survival; PFS= progression-free survival; TTLR, time to local relapse; TTDR, time to distant relapse; KRAS, kirsten rat sarcoma; OS, overall survival; PS, performance status; HR, hazard ratio; IMRT, intensity-modulated radiation therapy; CRT, chemoradiotherapy; PD-L1, programmed death-ligand 1; EGFR, epithelial growth factor receptor; BRAF, v-Raf murine sarcoma viral oncogene homolog B; ALK, anaplastic lymphoma kinase; STK11, serine/threonine kinase 11; RET, rearranged during transfection; TP53, tumor protein 53; p-value considered statistically significant was less than 0.05.

Glossary

ALK: anaplastic lymphoma kinase

BRAF: v-Raf murine sarcoma viral oncogene homolog B

CR: complete response

CRT: chemoradiotherapy

DCR: disease control rate

EGFR: epithelial growth factor receptor

HER2: human epidermal growth factor receptor 2

HR: hazard ratio

ICI: immune checkpoint inhibitor

IMRT: intensity-modulated radiation therapy

KL: co-mutation KRAS/STK11

KP: co-mutation KRAS/TP53

KRAS: Kirsten rat sarcoma

KRASm: KRAS mutation

KRASm G12C: KRAS G12C mutation

KRASm non-G12C: KRAS mutations excluding KRAS G12C mutation

KRASwt: KRAS wild-type

MET: mesenchymal epithelial transition

NGS: next-generation sequencing

NOS: not otherwise specified

NSCLC: non-small cell lung cancer

ORR: objective response rate

OS: overall survival

PCR: polymerase chain reaction

PD-L1: programmed death-ligand 1

PET: positron emission tomography

PFS: progression-free survival

PIK3CA: phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic

PR: partial response

RET: rearranged during transfection

ROS-1: c-ROS oncogene 1

STK11: serine/threonine kinase 11

TP53: tumor protein 53

TTDR: time to distant relapse

TTLR: time to local relapse

VEGF: vascular endothelial growth factor

References

1. 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) 71:209–49. doi: 10.3322/caac.21660

PubMed Abstract | Crossref Full Text | Google Scholar

2. Spigel DR, Faivre-Finn C, Gray JE, Vicente D, Planchard D, Paz-Ares L, et al. Five-year survival outcomes from the PACIFIC trial: durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. J Clin Oncol. (2022) 40:1301–11. doi: 10.1200/JCO.21.01308

PubMed Abstract | Crossref Full Text | Google Scholar

3. Herbst RS, Morgensztern D, and Boshoff C. The biology and management of non-small cell lung cancer. Nature. (2018) 553:446–54. doi: 10.1038/nature25183

PubMed Abstract | Crossref Full Text | Google Scholar

4. Prior IA, Hood FE, and Hartley JL. The frequency of ras mutations in cancer. Cancer Res. (2020) 80:2969–74. doi: 10.1158/0008-5472.CAN-19-3682

PubMed Abstract | Crossref Full Text | Google Scholar

5. Timar J and Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev. (2020) 39:1029–38. doi: 10.1007/s10555-020-09915-5

PubMed Abstract | Crossref Full Text | Google Scholar

6. Prior IA, Lewis PD, and Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer Res. (2012) 72:2457–67. doi: 10.1158/0008-5472.CAN-11-2612

PubMed Abstract | Crossref Full Text | Google Scholar

7. Judd J, Abdel Karim N, Khan H, Naqash AR, Baca Y, Xiu J, et al. Characterization of KRAS mutation subtypes in non-small cell lung cancer. Mol Cancer Ther. (2021) 20:2577–84. doi: 10.1158/1535-7163.MCT-21-0201

PubMed Abstract | Crossref Full Text | Google Scholar

8. Skoulidis F and Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer. (2019) 19:495–509. doi: 10.1038/s41568-019-0179-8

PubMed Abstract | Crossref Full Text | Google Scholar

9. Dogan S, Shen R, Ang DC, Johnson ML, D’Angelo SP, Paik PK, et al. Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRAS-mutant cancers. Clin Cancer Res. (2012) 18:6169–77. doi: 10.1158/1078-0432.CCR-11-3265

PubMed Abstract | Crossref Full Text | Google Scholar

10. 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) 14:876–89. doi: 10.1016/j.jtho.2019.01.020

PubMed Abstract | Crossref Full Text | Google Scholar

11. Wankhede D, Bontoux C, Grover S, and Hofman P. Prognostic role of KRAS G12C mutation in non-small cell lung cancer: A systematic review and meta-analysis. Diagnostics. (2023) 13:3043. doi: 10.3390/diagnostics13193043

PubMed Abstract | Crossref Full Text | Google Scholar

12. Finn SP, Addeo A, Dafni U, Thunnissen E, Bubendorf L, Madsen LB, et al. Prognostic impact of KRAS G12C mutation in patients with NSCLC: results from the european thoracic oncology platform lungscape project. J Thorac Oncol Off Publ Int Assoc Study Lung Cancer. (2021) 16:990–1002. doi: 10.1016/j.jtho.2021.02.016

PubMed Abstract | Crossref Full Text | Google Scholar

13. Frost MG, Jensen KJ, Gotfredsen DR, Sørensen AMS, Ankarfeldt MZ, Louie KS, et al. KRAS G12C mutated advanced non-small cell lung cancer (NSCLC): Characteristics, treatment patterns and overall survival from a Danish nationwide observational register study. Lung Cancer Amst Neth. (2023) 178:172–82. doi: 10.1016/j.lungcan.2023.02.021

PubMed Abstract | Crossref Full Text | Google Scholar

14. Sebastian M, Eberhardt WEE, Hoffknecht P, Metzenmacher M, Wehler T, Kokowski K, et al. KRAS G12C-mutated advanced non-small cell lung cancer: A real-world cohort from the German prospective, observational, nation-wide CRISP Registry (AIO-TRK-0315). Lung Cancer Amst Neth. (2021) 154:51–61. doi: 10.1016/j.lungcan.2021.02.005

PubMed Abstract | Crossref Full Text | Google Scholar

15. Gurtner K, Kryzmien Z, Koi L, Wang M, Benes CH, Hering S, et al. Radioresistance of KRAS/TP53-mutated lung cancer can be overcome by radiation dose escalation or EGFR tyrosine kinase inhibition in vivo. Int J Cancer. (2020) 147:472–7. doi: 10.1002/ijc.32598

PubMed Abstract | Crossref Full Text | Google Scholar

16. Arrieta O, Ramírez-Tirado LA, Caballé-Perez E, Mejia-Perez A, Zatarain-Barrón ZL, Cardona AF, et al. Response rate of patients with baseline brain metastases from recently diagnosed non-small cell lung cancer receiving radiotherapy according to EGFR, ALK and KRAS mutation status. Thorac Cancer. (2020) 11:1026–37. doi: 10.1111/1759-7714.13359

PubMed Abstract | Crossref Full Text | Google Scholar

17. Hu J, Zhang Z, Zhao L, Li L, Zuo W, and Han L. High expression of RAD51 promotes DNA damage repair and survival in KRAS-mutant lung cancer cells. BMB Rep. (2019) 52:151–6. doi: 10.5483/BMBRep.2019.52.2.213

PubMed Abstract | Crossref Full Text | Google Scholar

18. Wang M, Han J, Marcar L, Black J, Liu Q, Li X, et al. Radiation resistance in KRAS-mutated lung cancer is enabled by stem-like properties mediated by an osteopontin-EGFR pathway. Cancer Res. (2017) 77:2018–28. doi: 10.1158/0008-5472.CAN-16-0808

PubMed Abstract | Crossref Full Text | Google Scholar

19. Yagishita S, Horinouchi H, Sunami KS, Kanda S, Fujiwara Y, Nokihara H, et al. Impact of KRAS mutation on response and outcome of patients with stage III non-squamous non-small cell lung cancer. Cancer Sci. (2015) 106:1402–7. doi: 10.1111/cas.12740

PubMed Abstract | Crossref Full Text | Google Scholar

20. de Langen AJ, Johnson ML, Mazieres J, Dingemans AC, Mountzios G, Pless M, et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: a randomised, open-label, phase 3 trial. Lancet. (2023) 401:733–46. doi: 10.1016/S0140-6736(23)00221-0

PubMed Abstract | Crossref Full Text | Google Scholar

21. Jänne PA, Riely GJ, Gadgeel SM, Heist RS, Ou SI, Pacheco JM, et al. Adagrasib in non-small-cell lung cancer harboring a KRASG12C mutation. N Engl J Med. (2022) 387:120–31. doi: 10.1056/NEJMoa2204619

PubMed Abstract | Crossref Full Text | Google Scholar

22. Reck M, Carbone DP, Garassino M, and Barlesi F. Targeting KRAS in non-small-cell lung cancer: recent progress and new approaches. Ann Oncol. (2021) 32:1101–10. doi: 10.1016/j.annonc.2021.06.001

PubMed Abstract | Crossref Full Text | Google Scholar

23. Meng D, Yuan M, Li X, Chen L, Yang J, Zhao X, et al. Prognostic value of K-RAS mutations in patients with non-small cell lung cancer: a systematic review with meta-analysis. Lung Cancer Amst Neth. (2013) 81:1–10. doi: 10.1016/j.lungcan.2013.03.019

PubMed Abstract | Crossref Full Text | Google Scholar

24. Barsouk A, Friedes C, Iocolano M, Doucette A, Cohen RB, Robinson KW, et al. Plunging into the PACIFIC: outcomes of patients with unresectable KRAS-mutated non-small cell lung cancer following definitive chemoradiation and durvalumab consolidation. Clin Lung Cancer. (2024) 25:e161–71. doi: 10.1016/j.cllc.2023.12.009

PubMed Abstract | Crossref Full Text | Google Scholar

25. Villaruz LC, Socinski MA, Cunningham DE, Chiosea SI, Burns TF, Siegfried JM, et al. The prognostic and predictive value of KRAS oncogene substitutions in lung adenocarcinoma. Cancer. (2013) 119:2268–74. doi: 10.1002/cncr.28039

PubMed Abstract | Crossref Full Text | Google Scholar

26. Wahl SGF, Dai HY, Emdal EF, Berg T, Halvorsen TO, Ottestad AL, et al. The prognostic effect of KRAS mutations in non-small cell lung carcinoma revisited: A norwegian multicentre study. Cancers. (2021) 13:4294. doi: 10.3390/cancers13174294

PubMed Abstract | Crossref Full Text | Google Scholar

27. Cui W, Franchini F, Alexander M, Officer A, Wong HL, IJzerman M, et al. Real world outcomes in KRAS G12C mutation positive non-small cell lung cancer. Lung Cancer Amst Neth. (2020) 146:310–7. doi: 10.1016/j.lungcan.2020.06.030

PubMed Abstract | Crossref Full Text | Google Scholar

28. 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:e2215589. doi: 10.1001/jamanetworkopen.2022.15589

PubMed Abstract | Crossref Full Text | Google Scholar

29. Guo MZ, Murray JC, Ghanem P, Voong KR, Hales RK, Ettinger D, et al. Definitive chemoradiation and durvalumab consolidation for locally advanced, unresectable KRAS-mutated non-small cell lung cancer. Clin Lung Cancer. (2022) 23:620–9. doi: 10.1016/j.cllc.2022.08.002

PubMed Abstract | Crossref Full Text | Google Scholar

30. 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 Off J Am Assoc Cancer Res. (2017) 23:3012–24. doi: 10.1158/1078-0432.CCR-16-2554

PubMed Abstract | Crossref Full Text | Google Scholar

31. Papillon-Cavanagh S, Doshi P, Dobrin R, Szustakowski J, and Walsh AM. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort. ESMO Open. (2020) 5:e000706. doi: 10.1136/esmoopen-2020-000706

PubMed Abstract | Crossref Full Text | Google Scholar

32. Cao H, Ma Z, Li Y, Zhang Y, and Chen H. Prognostic value of KRAS G12C mutation in lung adenocarcinoma stratified by stages and radiological features. J thorac cardiovasc surg. (2023), S0022–5223(23)00361-6. doi: 10.1016/j.jtcvs.2023.04.037

PubMed Abstract | Crossref Full Text | Google Scholar

33. Arbour KC, Rizvi H, Plodkowski AJ, Hellmann MD, Knezevic A, Heller G, et al. Treatment outcomes and clinical characteristics of patients with KRAS-G12C-mutant non-small cell lung cancer. Clin Cancer Res. (2021) 27:2209–15. doi: 10.1158/1078-0432.CCR-20-4023

PubMed Abstract | Crossref Full Text | Google Scholar

34. Li D, Zhu X, Wang H, and Li N. Association between PD-L1 expression and driven gene status in NSCLC: A meta-analysis. Eur J Surg Oncol J Eur Soc Surg Oncol Br Assoc Surg Oncol. (2017) 43:1372–9. doi: 10.1016/j.ejso.2017.02.008

PubMed Abstract | Crossref Full Text | Google Scholar

35. Peters S, Ortega Granados ALO, de Marinis F, Lo Russo G, Schenker M, Arriola E, et al. KRYSTAL-7: Efficacy and safety of adagrasib with pembrolizumab in patients with treatment-naïve, advanced non small cell lung cancer (NSCLC) harboring a KRASG12C mutation. ESMO. (2023) 34(2):S1309–10. doi: 10.1016/j.annonc.2023.10.066

Crossref Full Text | Google Scholar

36. Burns TF, Dragnev KH, Fujiwara Y, Murciano-Goroff YR, and Lee DH. Efficacy and safety of olomorasib (LY3537982), a second-generation KRAS G12C inhibitor (G12Ci), in combination with pembrolizumab in patients with KRAS G12C-mutant advanced NSCLC. J Clin Oncol. (2024) 42(23_suppl). doi: 10.1200/JCO.2024.42.16_suppl.8510

Crossref Full Text | Google Scholar

37. Mok TSK, Yao W, Duruisseaux M, Doucet L, Martínez AA, Gregorc V, et al. KRYSTAL-12: Phase 3 study of adagrasib versus docetaxel in patients with previously treated advanced/metastatic non-small cell lung cancer (NSCLC) harboring a KRASG12C mutation. J Clin Oncol. (2024) 42(17_suppl). doi: 10.1200/JCO.2024.42.17_suppl.LBA8509

Crossref Full Text | Google Scholar

38. Lu S, Kato T, Dong X, Ahn MJ, Quang LV, Soparattanapaisarn N, et al. Osimertinib after chemoradiotherapy in stage III EGFR-mutated NSCLC. N Engl J Med. (2024) 391, 7. doi: 10.1056/NEJMoa2402614

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: non-small cell lung cancer, KRAS mutant, KRAS G12C mutation, chemoradiotherapy, locally advance disease

Citation: Cabon J, Lerouge D, Thureau S, Balosso J, Guigo M, Gervais R, Dubos C, Dô P, Demontrond P, Curcio H, Deshayes S, Madelaine J, Leite Ferreira D, Jacson L, Lecrosnier K, Chevalier F, Guisier F, Dantoing E, Leconte A, Christy F and Césaire M (2025) Impact of KRAS G12C mutation on the efficacy of chemoradiotherapy in patients with unresectable stage II or III non-small cell lung cancer. Front. Oncol. 15:1675825. doi: 10.3389/fonc.2025.1675825

Received: 29 July 2025; Accepted: 10 November 2025; Revised: 01 November 2025;
Published: 27 November 2025.

Edited by:

Chukwuka Eze, Ludwig Maximilian University of Munich, Germany

Reviewed by:

Mohamed Shanshal, Mayo Clinic, United States
Zsuzsanna Orosz, University of Debrecen, Hungary
Bohao Liu, The First Affiliated Hospital of Xi’an Jiaotong University, China

Copyright © 2025 Cabon, Lerouge, Thureau, Balosso, Guigo, Gervais, Dubos, Dô, Demontrond, Curcio, Deshayes, Madelaine, Leite Ferreira, Jacson, Lecrosnier, Chevalier, Guisier, Dantoing, Leconte, Christy and Césaire. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jean Cabon, amVhbi5jYWJvbjEwQGdtYWlsLmNvbQ==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.