Objective

We hypothesized that driver mutations in epidermal growth factor receptor (EGFR) are associated with decreased pathologic response to neoadjuvant chemoradiation (NA-ChRT) in locally advanced non-small cell lung cancer (LA-NSCLC).

Methods

Patients with Stage IIB-IIIA NSCLC treated with NA-ChRT, completion surgery, and underwent molecular profile testing were identified in a lung cancer database. Pathologic response was quantified using: (i) major pathologic response (MPR), (ii) complete pathologic response (pCR), and (iii) mean residual viable tumor cells (MRTC). Two groups were formed based on the presence or absence of driver mutations. Clinical and pathological correlations between the groups were studied.

Results

Forty-seven patients underwent tumor molecular profile testing, NA-ChRT, and completion surgery. Compared to the no-driver mutation group, the driver mutation group had lower MPR (23% vs 71%, p = 0.003), pCR (0% vs 26%, p = 0.02), and higher MRTC (43.4% vs 15.8%, p = 0.009). Univariate analysis showed an increased MPR rate for smokers, squamous cell histology, ChRT-surgery interval >65 days, and no-driver mutations. Multivariate analysis showed that only no-driver mutations (OR 0.39, p = 0.02) remained significant for MPR. PD-L1 status did not affect MPR. At 2 years, the driver mutation group had lower rates of local control (Hazard ration [HR] 0.67, p = 0.17) and disease-free survival (HR 0.5, p = 0.001). Overall survival was similar for both groups (HR = 1.04, p = 0.86).

Conclusion

Following 60 Gray NA-ChRT, tumors with a driver mutation had lower MPR and pCR rates than tumors without a driver mutation. PD-L1 was not associated with tumor regression.

Advances in knowledge

Patients with resectable LA-NSCLC and an EGFR driver mutation treated with neoadjuvant-ChRT and completion surgery have reduced pathologic regression, lower local control rates, and shorter disease-free survival than patients without a driver mutation. Evaluation of molecular testing should be introduced in LA-NSCLC intended for prognostication and treatment decisions.

Introduction

Epidermal growth factor receptor (EGFR/ErbB1) driver mutations are valuable biomarkers for predicting response to tyrosine kinase inhibitors (TKIs) in patients with metastatic lung adenocarcinoma.1,2 There is indirect evidence suggesting that the presence of EGFR/ErbB1 driver mutations may also offer prognostic information for patients with locally advanced non-small cell lung cancer (LA-NSCLC) treated with chemoradiation (ChRT).3 Evaluating the effects of TKIs in patients with LA-NSCLC treated with ChRT alone has been limited since molecular profiling of the tumor prior to starting treatment has not been mandatory.4,5 A recent analysis of the PACIFIC trial showed that patients with unresectable LA-NSCLC treated with adjuvant durvalumab following ChRT had improved disease-free survival (DFS) and overall survival (OS), suggested that patients with EGFR mutations may benefit less from treatment with adjuvant durvalumab than patients without EGFR mutations.6 However, since EGFR mutations were present in only 6% of patients enrolled in this trial, these results should be interpreted cautiously. Currently, most patients with unresectable LA-NSCLC are treated with definitive ChRT followed by adjuvant durvalumab without first testing for the presence of driver mutations.4–7

Since patients with driver mutations may benefit from including targeted therapies into their therapeutic regime, there is growing interest in extending TKIs to patients with LA-NSCLC who harbor EGFR mutations. Prior studies investigating adjuvant cetuximab or erlotinib/gefitinib failed to improve OS in patients with LA-NSCLC and driver mutations.8–10 More recently, the phase III ADAURA trial showed the use of adjuvant osimertinib for 3 years in patients with completely resected, EGFR mutated, Stage IB-IIIA NSCLC was associated with improved DFS compared to treatment with placebo (90% vs 44%, p < 0.001). Despite the use of adjuvant platinum-based chemotherapy (ChT) in 26% of patients with Stage IB disease and 76% of patients with Stage II-IIIA disease in addition to osimertinib, the improvement in DFS associated with the use of osimertinib compels further investigation with targeted therapies.11,12 Currently, to improve outcomes in patients with EGFR mutations, the phase III NEOADUARA trial is evaluating the use of neoadjuvant osimertinib in addition to adjuvant osimertinib in patients with resectable NSCLC,13 and the phase III LAURA trial is enrolling patients with Stage III unresectable LA-NSCLC to evaluate the use of osimertinib following completion of definitive ChRT.14

Zens et al15 and other authors suggested that the amount of pathologic regression in surgical specimens following NA-ChT or NA-ChRT in LA-NSCLC is an important prognostic factor associated with improved DFS and OS.16–19 Pataer et al showed in 192 NSCLC patients who received NA-ChT followed by surgery that both surgical pathologic stage and the presence of a major pathologic regression (MPR, ≤10% viable tumor cells) were associated with improved OS and DFS. Other authors concluded the same, making MPR a strong prognosticator for OS and DFS in patients with NSCLC.20–22 We hypothesized that driver mutations are associated with reduced tumor regression in surgical specimens following NA-ChRT.

To evaluate the effect of driver mutations on pathologic regression, we utilized a unique cohort of patients with resectable LA-NSCLC treated according to our institutional protocol with NA-ChRT followed by completion surgery (references blinded). Our primary end point was pathologic regression. Secondary end points were local control (LC), DFS, and OS. We correlated both primary and secondary end points to the presence or absence of EGFR mutations, PD-L1 status, and rare mutations.

Methods

Patients

We conducted a retrospective analysis of a database to identify patients with resectable Stage IIB-III NSCLC treated with NA-ChRT followed by pre-planned completion surgery that underwent tumor molecular profile testing at Sheba Medical Center between 2012 and 2021. Exclusion criteria for this analysis were unresectable disease, local or distant progression of disease during or after completion of ChRT, absence of molecular profile testing, radiation dose <50 Gy or ≥72 Gray (Gy), or non-concurrent ChRT. We excluded patients who underwent salvage surgery for progressive disease from this analysis.

Variables collected included age, sex, smoking history (current, past-smoker, never-smoker), clinical stage, gross tumor volume (GTV), lymph node volume, histology (adenocarcinoma, squamous cell carcinoma, and other histology), and tumor location (upper, middle, or lower lobes). We defined total tumor volume as the combined GTV and lymph node volumes. Treatment variables collected were the type of NA-ChT administered, radiation dose, radiation technique, use of adaptive replanning during the treatment course, use of durvalumab prior to surgery, the time interval between completion of NA-ChRT and surgery, and type of surgical procedure.

Molecular testing

We performed molecular testing to identify genetic alterations in tumor DNA; on the primary tumor or lymph nodes before starting ChRT, on lesions resected after ChRT and surgery, or on metastatic lesions. Kits used to determine the biomarkers changed during the time frame of this study, and based on the change in the standard testing used during different periods included EGFR testing using INFINITI Analyze (Auto Genomics, Carlsbad, CA)23 till 2018. After March 2018, Oncomine (Thermo Fisher Scientific, Waltham, MA), a next-generation sequencing test kit that analyzes for the presence of 22 genes simultaneously, was used to test for EGFR, ALK, ERBB2, ERBB4, FGFR1, FGFR2, FGFR3, MET, DDR2, KRAS, PIK3CA, BRAF, AKT1, PTEN, NRAS, MAP2K1, STK11, NOTCH1, CTNNB1, SMAD4, FBXW7 and TP53.24,25 Cases were assigned and analyzed according to the dominant genetic alteration, defined as the genetic alteration with the highest percentage of expression when several genetic alterations were detected.

PD-L1 expression was tested on archived tumor samples and measured by immunohistochemistry (Ventana Medical Systems, Oro Valley, AZ). The percentage of PD-L1 immunohistochemistry (IHC) staining was recorded from the pathologic specimens either before or after NA-ChRT and graded into three groups; negative (<1% positive staining), weakly positive (1–49%), and strongly positive (≥50%).26

Treatment

Since 2012, the practice at Sheba Medical Center for resectable LA-NSCLC was to deliver NA-ChRT using platinum-based doublet ChT concomitantly with RT to a dose of 60 Gray (Gy), followed by restaging and then completion surgery for cases without local or distant progression. The NA-ChRT and surgical treatment protocols utilized at our center have been reported previously.27 Since 2016, RT delivery protocol used volumetric arc therapy (VMAT, Varian Inc. Palo Alto, CA), with daily image-guided radiation therapy (IGRT) using cone beam computerized tomography (CBCT). As shown previously, using VMAT or three-dimensional conformal radiation therapy (3DCRT) planning resulted in similar rates of pathologic regression,28 thus both techniques were included.

Since the introduction of durvalumab, patients with borderline resectable stage-III disease, whose extent of disease was initially considered borderline for surgery, were offered the option of receiving adjuvant durvalumab. After repeated CT imaging and reevaluation, the multidisciplinary team decided to discontinue the durvalumab and proceed with completion surgery.

Pathologic response

Pathologic response to NA-ChRT was assessed from surgical specimens and quantified using three measures of pathologic response: (i) major pathologic response (MPR ≤10% viable tumor cells) vs no MPR (>10% tumor viable tumor cells), (ii) complete pathologic responses (pCR) and (iii) the percentage of mean residual viable tumor cells (MRTC). The pathologists interpreting the specimens were unaware of the presence or absence of driver mutations. Figure 1 shows examples of treatment effects: pCR, MPR, and residual disease.

(a–c): Example pathology slides of lobectomies after chemoradiation showing treatment effect: (a) pCR pathologic compleate response, (b) MPR major pathologic regression and (c) Residual disease.
Figure 1.

(ac): Example pathology slides of lobectomies after chemoradiation showing treatment effect: (a) pCR pathologic compleate response, (b) MPR major pathologic regression and (c) Residual disease.

Follow-up and clinical outcome

Patient follow-up examinations and imaging studies with CT or fludeoxyglucose (FDG) positron emission tomography (PET)-CT were done based on the standard of care, usually at 3 month intervals in the first-year post-surgery and then at 4–6 month intervals or as clinically indicated. We recorded the first recurrence as a local, regional, or distant failure.

Statistical analysis

Descriptive statistics used to assess patient and treatment parameters included proportions, means, and standard deviations. We compared variables between groups using a χ2 test, unpaired t-test (for age), Wilcoxon rank-sum (Mann–Whitney) test, Kruskal–Wallis for continuous variables not normally distributed, and Spearman’s rank test to correlate MRTC with continuous variables. We conducted univariate and multivariate logistic regression to predict the odds ratio (OR) for MPR. Patients were sorted into groups based on the presence or absence of driver mutations and subdivided based on surgical specimens’ pathologic response. We excluded rare mutations represented by only a single case from the regression models. Multivariate analysis (MVA) models included variables with a p-value of < 0.2 in the univariate analysis (UVA). We excluded variables correlating with EGFR with significant (p ≤ 0.05) correlation coefficients from the MVA. OS was recorded from the start of NA-ChRT till death or censured at the last time the patient was known to be alive. Local and distant progression were recorded from patients' medical records or review of CT or FDG PET-CT imaging studies and was calculated from the initiation of NA-ChRT till progression or censure at the last evaluation. Time-to-event data were analyzed using the Kaplan–Meier method. We excluded driver mutations represented by a single case from the Kaplan–Meier analysis. We used hazard ratios (HRs) to compare local failure, distant failure, and survival rates. A p-value ≤ 0.05 was considered statistically significant. We used STATA v. 13 (StataCorp. 2013, Stata Statistical Software: Release 13. College Station, TX: StataCorp LP) for all statistical analysis.

Results

During the study period, 107 patients with resectable LA-NSCLC were treated with NA-ChRT followed by surgery. Tumor molecular profile testing was available in 47 patients. Patients not undergoing testing for mutations (n = 60) were excluded from this analysis.

The patient characteristics, shown in Table 1, are subdivided into two groups based on tumor molecular profile testing results. The driver mutation group included patients with alterations in recognized drivers; EGFR (n = 13), BRAF (n = 1), RET (n = 1), and ROS1 (n = 1). The no-driver mutation group included all patients with no mutation identified (n = 17) as well as patients with mutation or aberration in FGFR (n = 2), KRAS (n = 10), ERBB4 (n = 1), or AKT (n = 1). Although some of these genes may be considered driver mutations, we designated this group the ‘no-driver-mutation group’ since, besides KRAS, none were proven drivers in lung cancer. Patients with KRAS mutation have clinical characteristics similar to patients with no-driver mutations, justifying their inclusion within the ‘no-driver mutation’ group.29

Table 1.

Patient characteristics

Patient characteristicAll patientsNo-driver mutationDriver mutationp- value
Patient number473116
Age (mean, SD)61 (8.9)62 (8.6)59 (9.4)0.25
Sex:0.58
 Female18117
 Male29209
Smoking:0.00
 Never15411
 Past523
 Current27261
Histology:0.016
 Adenocarcinoma351916
 Squamous cell carcinoma550
 Other770
Stage (AJCC 7th ed.):0.58
 II541
 III412615
 IV110
Tumor location:0.8
 Upper /middle lobes342212
 Lower lobes1394
Chemotherapy:0.14
 Cisplatin-vinorelbine1073
 Carboplatin-paclitaxel271512
 Platinum-etoposide770
 Cisplatin-pemetrexed321
Neoadjuvant durvalumab:0.06
 No412516
 Yes660
Surgery type:0.25
 Chest wall resection + lobectomy440
 Lobectomy342212
 Pneumonectomy853
PDL1 IHC (total 34) percent (mean, (SD)29.1(33)29.4 (35)28.7 (30.3)0.95
Negative1192
Weak positive (<50%)12660.24
Strong positive (≥50%)1183
Radiation dose: (Mean, SD)61 (5)61.2 (5.5)61 (5)0.9
Planning technique:0.26
 three-dimensional fields (3DCRT)20155
 Volumetric arc (VMAT)271611
Adaptive volumetric: replanning0.13
 Yes1091
 No362115
 Missing010
Total GTV volume primary + nodes (cm3), (mean, SD)140 (119)172 (132)81 (54.5)0.016
GTV, cm3, (mean, SD)110 (123)143 (137)48 (47.5)0.047
Lymph nodes volume, cm3 (mean, SD)30.7 (32.6)29.6 (37)32.6 (22)0.12
Lung V20 (%) (mean, SD)23 (6)23.1 (6.5)23.4 (4.7)0.76
Mean lung dose (Gy) (mean, SD)13.6 (2.5)13.6 (3.5)13.6 (2.4)0.83
Lung V5 (%) (mean, SD)48.3 (14.6)48.4 (16)48.3 (11.6)0.95
Heart V45 (%) (mean, SD)6.5 (11.8)7.9 (13.4)4 (11.8)0.4
Mean heart dose (Gy) (mean, SD)9 (9.2)9.65 (9.6)8 (8.5)0.7
Spine max dose (Gy) (mean, SD)38.3 (11.7)37.5 (13.2)29.8 (8.3)0.88
PTV D95% (mean, SD)95.5 (3.1)95 (2.7)96.4 (3.7)0.87
Interval between radiation and surgery (days) (mean, SD) (including durvalumab)72.8 (44.7)81 (50) (n=31)56 (20)0.01
Interval between radiation and surgery (days) (mean, SD) (excluding durvalumab)61.8 (24) (n=25)56 (20)0.33
Interval between radiation and surgery (days) (mean, SD) (only durvalumab)163 (44) (n=6)56 (20)0.002
Patient characteristicAll patientsNo-driver mutationDriver mutationp- value
Patient number473116
Age (mean, SD)61 (8.9)62 (8.6)59 (9.4)0.25
Sex:0.58
 Female18117
 Male29209
Smoking:0.00
 Never15411
 Past523
 Current27261
Histology:0.016
 Adenocarcinoma351916
 Squamous cell carcinoma550
 Other770
Stage (AJCC 7th ed.):0.58
 II541
 III412615
 IV110
Tumor location:0.8
 Upper /middle lobes342212
 Lower lobes1394
Chemotherapy:0.14
 Cisplatin-vinorelbine1073
 Carboplatin-paclitaxel271512
 Platinum-etoposide770
 Cisplatin-pemetrexed321
Neoadjuvant durvalumab:0.06
 No412516
 Yes660
Surgery type:0.25
 Chest wall resection + lobectomy440
 Lobectomy342212
 Pneumonectomy853
PDL1 IHC (total 34) percent (mean, (SD)29.1(33)29.4 (35)28.7 (30.3)0.95
Negative1192
Weak positive (<50%)12660.24
Strong positive (≥50%)1183
Radiation dose: (Mean, SD)61 (5)61.2 (5.5)61 (5)0.9
Planning technique:0.26
 three-dimensional fields (3DCRT)20155
 Volumetric arc (VMAT)271611
Adaptive volumetric: replanning0.13
 Yes1091
 No362115
 Missing010
Total GTV volume primary + nodes (cm3), (mean, SD)140 (119)172 (132)81 (54.5)0.016
GTV, cm3, (mean, SD)110 (123)143 (137)48 (47.5)0.047
Lymph nodes volume, cm3 (mean, SD)30.7 (32.6)29.6 (37)32.6 (22)0.12
Lung V20 (%) (mean, SD)23 (6)23.1 (6.5)23.4 (4.7)0.76
Mean lung dose (Gy) (mean, SD)13.6 (2.5)13.6 (3.5)13.6 (2.4)0.83
Lung V5 (%) (mean, SD)48.3 (14.6)48.4 (16)48.3 (11.6)0.95
Heart V45 (%) (mean, SD)6.5 (11.8)7.9 (13.4)4 (11.8)0.4
Mean heart dose (Gy) (mean, SD)9 (9.2)9.65 (9.6)8 (8.5)0.7
Spine max dose (Gy) (mean, SD)38.3 (11.7)37.5 (13.2)29.8 (8.3)0.88
PTV D95% (mean, SD)95.5 (3.1)95 (2.7)96.4 (3.7)0.87
Interval between radiation and surgery (days) (mean, SD) (including durvalumab)72.8 (44.7)81 (50) (n=31)56 (20)0.01
Interval between radiation and surgery (days) (mean, SD) (excluding durvalumab)61.8 (24) (n=25)56 (20)0.33
Interval between radiation and surgery (days) (mean, SD) (only durvalumab)163 (44) (n=6)56 (20)0.002

Driver mutation group includes mutations in EGFR, RET, ROS and BRAF. GTV: gross tumor volume of the primary lesion, Lung V20: percent of lung receiving 20 Gray, Lung V5: percent of the lung receiving 5 Gray, Heart V45: percent of the heart receiving 45 Gray, PTV D95% the volume of the planning target volume receiving 95% of the dose. Programmed death-ligand one (PD-L1) Immunohistochemistry (IHC)

Table 1.

Patient characteristics

Patient characteristicAll patientsNo-driver mutationDriver mutationp- value
Patient number473116
Age (mean, SD)61 (8.9)62 (8.6)59 (9.4)0.25
Sex:0.58
 Female18117
 Male29209
Smoking:0.00
 Never15411
 Past523
 Current27261
Histology:0.016
 Adenocarcinoma351916
 Squamous cell carcinoma550
 Other770
Stage (AJCC 7th ed.):0.58
 II541
 III412615
 IV110
Tumor location:0.8
 Upper /middle lobes342212
 Lower lobes1394
Chemotherapy:0.14
 Cisplatin-vinorelbine1073
 Carboplatin-paclitaxel271512
 Platinum-etoposide770
 Cisplatin-pemetrexed321
Neoadjuvant durvalumab:0.06
 No412516
 Yes660
Surgery type:0.25
 Chest wall resection + lobectomy440
 Lobectomy342212
 Pneumonectomy853
PDL1 IHC (total 34) percent (mean, (SD)29.1(33)29.4 (35)28.7 (30.3)0.95
Negative1192
Weak positive (<50%)12660.24
Strong positive (≥50%)1183
Radiation dose: (Mean, SD)61 (5)61.2 (5.5)61 (5)0.9
Planning technique:0.26
 three-dimensional fields (3DCRT)20155
 Volumetric arc (VMAT)271611
Adaptive volumetric: replanning0.13
 Yes1091
 No362115
 Missing010
Total GTV volume primary + nodes (cm3), (mean, SD)140 (119)172 (132)81 (54.5)0.016
GTV, cm3, (mean, SD)110 (123)143 (137)48 (47.5)0.047
Lymph nodes volume, cm3 (mean, SD)30.7 (32.6)29.6 (37)32.6 (22)0.12
Lung V20 (%) (mean, SD)23 (6)23.1 (6.5)23.4 (4.7)0.76
Mean lung dose (Gy) (mean, SD)13.6 (2.5)13.6 (3.5)13.6 (2.4)0.83
Lung V5 (%) (mean, SD)48.3 (14.6)48.4 (16)48.3 (11.6)0.95
Heart V45 (%) (mean, SD)6.5 (11.8)7.9 (13.4)4 (11.8)0.4
Mean heart dose (Gy) (mean, SD)9 (9.2)9.65 (9.6)8 (8.5)0.7
Spine max dose (Gy) (mean, SD)38.3 (11.7)37.5 (13.2)29.8 (8.3)0.88
PTV D95% (mean, SD)95.5 (3.1)95 (2.7)96.4 (3.7)0.87
Interval between radiation and surgery (days) (mean, SD) (including durvalumab)72.8 (44.7)81 (50) (n=31)56 (20)0.01
Interval between radiation and surgery (days) (mean, SD) (excluding durvalumab)61.8 (24) (n=25)56 (20)0.33
Interval between radiation and surgery (days) (mean, SD) (only durvalumab)163 (44) (n=6)56 (20)0.002
Patient characteristicAll patientsNo-driver mutationDriver mutationp- value
Patient number473116
Age (mean, SD)61 (8.9)62 (8.6)59 (9.4)0.25
Sex:0.58
 Female18117
 Male29209
Smoking:0.00
 Never15411
 Past523
 Current27261
Histology:0.016
 Adenocarcinoma351916
 Squamous cell carcinoma550
 Other770
Stage (AJCC 7th ed.):0.58
 II541
 III412615
 IV110
Tumor location:0.8
 Upper /middle lobes342212
 Lower lobes1394
Chemotherapy:0.14
 Cisplatin-vinorelbine1073
 Carboplatin-paclitaxel271512
 Platinum-etoposide770
 Cisplatin-pemetrexed321
Neoadjuvant durvalumab:0.06
 No412516
 Yes660
Surgery type:0.25
 Chest wall resection + lobectomy440
 Lobectomy342212
 Pneumonectomy853
PDL1 IHC (total 34) percent (mean, (SD)29.1(33)29.4 (35)28.7 (30.3)0.95
Negative1192
Weak positive (<50%)12660.24
Strong positive (≥50%)1183
Radiation dose: (Mean, SD)61 (5)61.2 (5.5)61 (5)0.9
Planning technique:0.26
 three-dimensional fields (3DCRT)20155
 Volumetric arc (VMAT)271611
Adaptive volumetric: replanning0.13
 Yes1091
 No362115
 Missing010
Total GTV volume primary + nodes (cm3), (mean, SD)140 (119)172 (132)81 (54.5)0.016
GTV, cm3, (mean, SD)110 (123)143 (137)48 (47.5)0.047
Lymph nodes volume, cm3 (mean, SD)30.7 (32.6)29.6 (37)32.6 (22)0.12
Lung V20 (%) (mean, SD)23 (6)23.1 (6.5)23.4 (4.7)0.76
Mean lung dose (Gy) (mean, SD)13.6 (2.5)13.6 (3.5)13.6 (2.4)0.83
Lung V5 (%) (mean, SD)48.3 (14.6)48.4 (16)48.3 (11.6)0.95
Heart V45 (%) (mean, SD)6.5 (11.8)7.9 (13.4)4 (11.8)0.4
Mean heart dose (Gy) (mean, SD)9 (9.2)9.65 (9.6)8 (8.5)0.7
Spine max dose (Gy) (mean, SD)38.3 (11.7)37.5 (13.2)29.8 (8.3)0.88
PTV D95% (mean, SD)95.5 (3.1)95 (2.7)96.4 (3.7)0.87
Interval between radiation and surgery (days) (mean, SD) (including durvalumab)72.8 (44.7)81 (50) (n=31)56 (20)0.01
Interval between radiation and surgery (days) (mean, SD) (excluding durvalumab)61.8 (24) (n=25)56 (20)0.33
Interval between radiation and surgery (days) (mean, SD) (only durvalumab)163 (44) (n=6)56 (20)0.002

Driver mutation group includes mutations in EGFR, RET, ROS and BRAF. GTV: gross tumor volume of the primary lesion, Lung V20: percent of lung receiving 20 Gray, Lung V5: percent of the lung receiving 5 Gray, Heart V45: percent of the heart receiving 45 Gray, PTV D95% the volume of the planning target volume receiving 95% of the dose. Programmed death-ligand one (PD-L1) Immunohistochemistry (IHC)

Comparison between the groups showed that the driver mutation group consisted of adenocarcinomas, mostly never smokers, had smaller GTVs (p = 0.047), smaller total tumor volumes (p = 0.016), and did not receive durvalumab post-ChRT (p = 0.08).

The no-driver mutation group had both adeno- and squamous cell carcinomas, were primarily smokers, had larger GTVs, larger total tumor volumes, and included a few patients (n = 6) who received durvalumab prior to undergoing completion surgery. The time interval between the end of NA-ChRT and surgery was longer for the six cases treated with durvalumab prior to surgery (mean 163 days, SD 44) than cases in the no-driver mutation group that did not receive durvalumab (mean 61.8 days, SD 24), or the driver mutation group (mean 56 days, SD 20, p = 0.002) (Table 1).

Table 2A presents each identified biomarker and its response to therapy measured by MPR, pCR, and MRTC in the surgical specimens following NA-ChRT. The driver mutation group demonstrated low pCR, MPR, and high MRTC. The MRTC for this group was 43.4% for EGFR (n = 13), 50% for RET (n = 1), 50% for ROS1 (50%, n = 1), and 45% for BRAF (n = 1). Mutations represented by only a solitary case each (RET/ROS1/BRAF) were excluded from the driver mutation group for the UVA and MVA, leaving an EGFR-only group (n = 13) for analysis.

Table 2.

Pathologic response to chemo-radiation according to EGFR mutation

GroupingThe leading mutationMPRpCRMean residual tumor cells (%)
Driver mutationsEGFR N = 133/13 (23%)0/13 (0%)43.4% (SD35)
BRAF N = 10/1 (0%)0/1 (0%)45%
RET N = 10/1 (0%)0/1 (0%)50%
ROS N = 10/1 (0%)0/1 (0%)50%
No driver mutationsFGFR N = 22/2 (100%)2/2 (100%)0% (SD 0)
KRAS N = 108/10 (80%)4/10 (40%)13.7% (SD 23)
ERBB4 N = 11/1 (100%)0/1 (100%)5%
AKT amplification N = 11/1 (100%)0/1 (100%)9%
Negative for mutation N = 1711/17 (64.7%)3/17 (17.6%)19.9% (SD24)
Biomarker groupingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Driver mutations N = 163/16 (19%)0.00010/16 (0%)0.00444.4% (SD 31)0.001
EGFR only mutations N = 133/13 (23%)0.0030/13 (0%)0.0243.4% (SD 35)0.009
No driver mutations N = 3122/31 (71%)reference8/31 (26%)reference15.8% (SD 22.4)reference
GroupingThe leading mutationMPRpCRMean residual tumor cells (%)
Driver mutationsEGFR N = 133/13 (23%)0/13 (0%)43.4% (SD35)
BRAF N = 10/1 (0%)0/1 (0%)45%
RET N = 10/1 (0%)0/1 (0%)50%
ROS N = 10/1 (0%)0/1 (0%)50%
No driver mutationsFGFR N = 22/2 (100%)2/2 (100%)0% (SD 0)
KRAS N = 108/10 (80%)4/10 (40%)13.7% (SD 23)
ERBB4 N = 11/1 (100%)0/1 (100%)5%
AKT amplification N = 11/1 (100%)0/1 (100%)9%
Negative for mutation N = 1711/17 (64.7%)3/17 (17.6%)19.9% (SD24)
Biomarker groupingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Driver mutations N = 163/16 (19%)0.00010/16 (0%)0.00444.4% (SD 31)0.001
EGFR only mutations N = 133/13 (23%)0.0030/13 (0%)0.0243.4% (SD 35)0.009
No driver mutations N = 3122/31 (71%)reference8/31 (26%)reference15.8% (SD 22.4)reference

Abbreviations: Epidermal growth factor receptor (EGFR), Major pathologic response (MPR), Mean residual tumor cells (MRTC) Pathologic complete response (pCR). The driver mutations group include EGFR, RET, ROS1 and BRAF. The EGFR only group excluded RET, ROS and BRAF from the analysis. P values are for comparison of no- driver mutations to driver mutations and EGFR only mutations.

Table 2.

Pathologic response to chemo-radiation according to EGFR mutation

GroupingThe leading mutationMPRpCRMean residual tumor cells (%)
Driver mutationsEGFR N = 133/13 (23%)0/13 (0%)43.4% (SD35)
BRAF N = 10/1 (0%)0/1 (0%)45%
RET N = 10/1 (0%)0/1 (0%)50%
ROS N = 10/1 (0%)0/1 (0%)50%
No driver mutationsFGFR N = 22/2 (100%)2/2 (100%)0% (SD 0)
KRAS N = 108/10 (80%)4/10 (40%)13.7% (SD 23)
ERBB4 N = 11/1 (100%)0/1 (100%)5%
AKT amplification N = 11/1 (100%)0/1 (100%)9%
Negative for mutation N = 1711/17 (64.7%)3/17 (17.6%)19.9% (SD24)
Biomarker groupingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Driver mutations N = 163/16 (19%)0.00010/16 (0%)0.00444.4% (SD 31)0.001
EGFR only mutations N = 133/13 (23%)0.0030/13 (0%)0.0243.4% (SD 35)0.009
No driver mutations N = 3122/31 (71%)reference8/31 (26%)reference15.8% (SD 22.4)reference
GroupingThe leading mutationMPRpCRMean residual tumor cells (%)
Driver mutationsEGFR N = 133/13 (23%)0/13 (0%)43.4% (SD35)
BRAF N = 10/1 (0%)0/1 (0%)45%
RET N = 10/1 (0%)0/1 (0%)50%
ROS N = 10/1 (0%)0/1 (0%)50%
No driver mutationsFGFR N = 22/2 (100%)2/2 (100%)0% (SD 0)
KRAS N = 108/10 (80%)4/10 (40%)13.7% (SD 23)
ERBB4 N = 11/1 (100%)0/1 (100%)5%
AKT amplification N = 11/1 (100%)0/1 (100%)9%
Negative for mutation N = 1711/17 (64.7%)3/17 (17.6%)19.9% (SD24)
Biomarker groupingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Driver mutations N = 163/16 (19%)0.00010/16 (0%)0.00444.4% (SD 31)0.001
EGFR only mutations N = 133/13 (23%)0.0030/13 (0%)0.0243.4% (SD 35)0.009
No driver mutations N = 3122/31 (71%)reference8/31 (26%)reference15.8% (SD 22.4)reference

Abbreviations: Epidermal growth factor receptor (EGFR), Major pathologic response (MPR), Mean residual tumor cells (MRTC) Pathologic complete response (pCR). The driver mutations group include EGFR, RET, ROS1 and BRAF. The EGFR only group excluded RET, ROS and BRAF from the analysis. P values are for comparison of no- driver mutations to driver mutations and EGFR only mutations.

Mutations within the no-driver mutation group had higher rates of pCR, MPR, and lower MRTC. This group had low MRTC: 13.7% for KRAS (n = 10), 5% for ERBB4 (n = 1), 9% for AKT (n = 1), 0% for FGFR (n = 2), and 19.9% for patients who had no mutations identified (n = 17) on molecular testing. The no-driver mutation group was unchanged for the UVA and MVA. The no-driver mutation group was composed chiefly of cases with either KRAS mutation (n = 10) or no identifiable mutation (n = 17). These two subgroups of the no-driver-mutation group showed similar rates of pCR, MPR, and MRTC. Furthermore, Kaplan–Meier estimates of local control, DFS, and OS at 2 years for KRAS mutations and no-mutations showed no significant differences between cases with KRAS or no mutations identified (Figure S1 A–C). For KRAS vs. no mutations identified, LC at 2 years was 80% (95% CI 40–95) vs. 89% (95% CI 62–97) (p = 0.2), DFS was 60% (95% CI 25–83%) vs. 41% (95% CI 20–63%) (p = 0.7), and OS trended lower for KRAS: 70% (95% CI 33–89) vs. 90.5% (95% CI 67–97) (p = 0.18).

Comparison between the EGFR-only and no-driver mutation groups (Table 2B) showed the EGFR group had higher rates of MRTC (43.4% vs. 15.8%, p = 0.009) and lower rates of MPR (23% vs. 71%, p = 0.003) and pCR (0% vs. 26%, p = 0.02).

The UVA (Table 3) showed that the likelihood of MPR was increased for smokers (OR 0.34, p = 0.005), squamous cell histology (OR 0.53, p = 0.063), NA-ChRT surgery interval >65 days (OR 0.12, p = 0.005), and absence of an EGFR mutation (OR 0.35, p = 0.006). Cases receiving durvalumab before surgery trended towards a higher rate of MPR (83% vs 53%, OR 0.45, p = 0.16). PD-L1 status did not influence MPR.

Table 3.

Uni- and multivariate analysis predicting the odds ratio for MPR

VariableMPR N/TOTAL (%)UVAMVA
Odds ratio (95% CI)p- valueOdds ratio (95% CI)p- value
Age0.99 (0.95-1.04)0.78
Sex:0.5 (0.17-1.9)0.34
 Female n=188/18 (44%)
 Male n=2917/29 (58%)
Smokinga:0.34 (0.16-0.7)0.004
 Never n=153/15 (20%)
 Past n=53/5 (60%)
 Current N=2719/27 (70%)
Histologya:0.53 (0.3-1.01)0.054
 Squamous n=55/5 (100%)
 Adenocarcinoma n=3516/35 (46%)
 Other histology n=74/7 (57%)
Stage:0.3 (0.07-2.5)0.35
 II n=52/5 (40%)
 III n=4022/41 (53%)
 IV n=11/1 (100%)
Tumor volume:0.53 (0.16-1.7)0.28
 ≤115cc n=2512/25 (48%)
 >115cc n=2113/21 (62%)
Location of primary tumor:2.5 (0.6-9.8)0.181.7 (0.4-7.5)0.45
 Upper lobe n=3416/34 (48%)
 Lower lobe n=139/13 (69%)
Chemotherapy:
 Cisplatin-vinorelbine4/10 (40%)0.98 (0.63-1.5)0.92
 Carboplatin-paclitaxel Q1W13/24 (54%)
 Carboplatin-paclitaxel Q3W2/2 (100%)
 Cisplatin-etoposide6/7 (86%)
 Cisplatin-pemetrexed0/3 (0%)
Durvalumab:0.43 (0.15-1.3)0.150.7 (0.2-2.1)0.48
 No n=4120/41 (49%)
 Yes n=65/6 (83%)
Radiation dose:0.99 (0.3-3.4)0.99
 ≤55 Gy n=3217/32 (53%)
 >56 Gy n=158/15 (53%)
Radiation technique:0.8 (0.25-2.5)0.7
 3DCT n=2010/20 (50%)
 VMAT n=2715/27 (56%)
Breathing management:0.66 (0. 3-1.4)0.3
 DIBH n=83/8 (37%)
 FB n=3822/38 (58%)
Adaptive planning due to tumor shrinkage:2.9 (0.7-12)0.131.6 (0.37-6.6)0.53
 Yes n=107/10 (70%)
 No n=3617/36 (47%)
 No-EGFR n=3122/31 (71%)0.35 (0.16-0.7)0.0060.39 (0.18-0.86)0.02
 EGFR n=133/13 (23%)
PDL1 statusb:1 (0.43-2.3)1
 Negative n=116/11 (54%)
 Weak positive n=115/11 (45%)
 Strong positive n=116/11 (54%)
Time interval radiation to surgerya:0.12 (0.02-0.5)0.005
 ≤65 days n=3011/30 (36.7%)
 >65 days n=1714/17 (82%)
VariableMPR N/TOTAL (%)UVAMVA
Odds ratio (95% CI)p- valueOdds ratio (95% CI)p- value
Age0.99 (0.95-1.04)0.78
Sex:0.5 (0.17-1.9)0.34
 Female n=188/18 (44%)
 Male n=2917/29 (58%)
Smokinga:0.34 (0.16-0.7)0.004
 Never n=153/15 (20%)
 Past n=53/5 (60%)
 Current N=2719/27 (70%)
Histologya:0.53 (0.3-1.01)0.054
 Squamous n=55/5 (100%)
 Adenocarcinoma n=3516/35 (46%)
 Other histology n=74/7 (57%)
Stage:0.3 (0.07-2.5)0.35
 II n=52/5 (40%)
 III n=4022/41 (53%)
 IV n=11/1 (100%)
Tumor volume:0.53 (0.16-1.7)0.28
 ≤115cc n=2512/25 (48%)
 >115cc n=2113/21 (62%)
Location of primary tumor:2.5 (0.6-9.8)0.181.7 (0.4-7.5)0.45
 Upper lobe n=3416/34 (48%)
 Lower lobe n=139/13 (69%)
Chemotherapy:
 Cisplatin-vinorelbine4/10 (40%)0.98 (0.63-1.5)0.92
 Carboplatin-paclitaxel Q1W13/24 (54%)
 Carboplatin-paclitaxel Q3W2/2 (100%)
 Cisplatin-etoposide6/7 (86%)
 Cisplatin-pemetrexed0/3 (0%)
Durvalumab:0.43 (0.15-1.3)0.150.7 (0.2-2.1)0.48
 No n=4120/41 (49%)
 Yes n=65/6 (83%)
Radiation dose:0.99 (0.3-3.4)0.99
 ≤55 Gy n=3217/32 (53%)
 >56 Gy n=158/15 (53%)
Radiation technique:0.8 (0.25-2.5)0.7
 3DCT n=2010/20 (50%)
 VMAT n=2715/27 (56%)
Breathing management:0.66 (0. 3-1.4)0.3
 DIBH n=83/8 (37%)
 FB n=3822/38 (58%)
Adaptive planning due to tumor shrinkage:2.9 (0.7-12)0.131.6 (0.37-6.6)0.53
 Yes n=107/10 (70%)
 No n=3617/36 (47%)
 No-EGFR n=3122/31 (71%)0.35 (0.16-0.7)0.0060.39 (0.18-0.86)0.02
 EGFR n=133/13 (23%)
PDL1 statusb:1 (0.43-2.3)1
 Negative n=116/11 (54%)
 Weak positive n=115/11 (45%)
 Strong positive n=116/11 (54%)
Time interval radiation to surgerya:0.12 (0.02-0.5)0.005
 ≤65 days n=3011/30 (36.7%)
 >65 days n=1714/17 (82%)

Major pathologic regression (MPR), Gray (Gy), Three-dimensional conformal radiation therapy (3DCRT), Volumetric arc therapy (VMAT), Deep inspiratory breath hold (DIBH), Free breathing (FB), Epidermal growth factor receptor (EGFR), Chemoradiation (ChRT).

a

Smoking, histology, and time interval between ChRT and surgery correlated with EGFR mutation (smoking p < 0.0001, histology p = 0.013, time interval between ChRT and surgery p = 0.048). Thus, they were excluded from the MVA.

b

PDL1 status negative (< 1% positive staining); weak positive (1–49%) and strong positive (≥ 50%).

Table 3.

Uni- and multivariate analysis predicting the odds ratio for MPR

VariableMPR N/TOTAL (%)UVAMVA
Odds ratio (95% CI)p- valueOdds ratio (95% CI)p- value
Age0.99 (0.95-1.04)0.78
Sex:0.5 (0.17-1.9)0.34
 Female n=188/18 (44%)
 Male n=2917/29 (58%)
Smokinga:0.34 (0.16-0.7)0.004
 Never n=153/15 (20%)
 Past n=53/5 (60%)
 Current N=2719/27 (70%)
Histologya:0.53 (0.3-1.01)0.054
 Squamous n=55/5 (100%)
 Adenocarcinoma n=3516/35 (46%)
 Other histology n=74/7 (57%)
Stage:0.3 (0.07-2.5)0.35
 II n=52/5 (40%)
 III n=4022/41 (53%)
 IV n=11/1 (100%)
Tumor volume:0.53 (0.16-1.7)0.28
 ≤115cc n=2512/25 (48%)
 >115cc n=2113/21 (62%)
Location of primary tumor:2.5 (0.6-9.8)0.181.7 (0.4-7.5)0.45
 Upper lobe n=3416/34 (48%)
 Lower lobe n=139/13 (69%)
Chemotherapy:
 Cisplatin-vinorelbine4/10 (40%)0.98 (0.63-1.5)0.92
 Carboplatin-paclitaxel Q1W13/24 (54%)
 Carboplatin-paclitaxel Q3W2/2 (100%)
 Cisplatin-etoposide6/7 (86%)
 Cisplatin-pemetrexed0/3 (0%)
Durvalumab:0.43 (0.15-1.3)0.150.7 (0.2-2.1)0.48
 No n=4120/41 (49%)
 Yes n=65/6 (83%)
Radiation dose:0.99 (0.3-3.4)0.99
 ≤55 Gy n=3217/32 (53%)
 >56 Gy n=158/15 (53%)
Radiation technique:0.8 (0.25-2.5)0.7
 3DCT n=2010/20 (50%)
 VMAT n=2715/27 (56%)
Breathing management:0.66 (0. 3-1.4)0.3
 DIBH n=83/8 (37%)
 FB n=3822/38 (58%)
Adaptive planning due to tumor shrinkage:2.9 (0.7-12)0.131.6 (0.37-6.6)0.53
 Yes n=107/10 (70%)
 No n=3617/36 (47%)
 No-EGFR n=3122/31 (71%)0.35 (0.16-0.7)0.0060.39 (0.18-0.86)0.02
 EGFR n=133/13 (23%)
PDL1 statusb:1 (0.43-2.3)1
 Negative n=116/11 (54%)
 Weak positive n=115/11 (45%)
 Strong positive n=116/11 (54%)
Time interval radiation to surgerya:0.12 (0.02-0.5)0.005
 ≤65 days n=3011/30 (36.7%)
 >65 days n=1714/17 (82%)
VariableMPR N/TOTAL (%)UVAMVA
Odds ratio (95% CI)p- valueOdds ratio (95% CI)p- value
Age0.99 (0.95-1.04)0.78
Sex:0.5 (0.17-1.9)0.34
 Female n=188/18 (44%)
 Male n=2917/29 (58%)
Smokinga:0.34 (0.16-0.7)0.004
 Never n=153/15 (20%)
 Past n=53/5 (60%)
 Current N=2719/27 (70%)
Histologya:0.53 (0.3-1.01)0.054
 Squamous n=55/5 (100%)
 Adenocarcinoma n=3516/35 (46%)
 Other histology n=74/7 (57%)
Stage:0.3 (0.07-2.5)0.35
 II n=52/5 (40%)
 III n=4022/41 (53%)
 IV n=11/1 (100%)
Tumor volume:0.53 (0.16-1.7)0.28
 ≤115cc n=2512/25 (48%)
 >115cc n=2113/21 (62%)
Location of primary tumor:2.5 (0.6-9.8)0.181.7 (0.4-7.5)0.45
 Upper lobe n=3416/34 (48%)
 Lower lobe n=139/13 (69%)
Chemotherapy:
 Cisplatin-vinorelbine4/10 (40%)0.98 (0.63-1.5)0.92
 Carboplatin-paclitaxel Q1W13/24 (54%)
 Carboplatin-paclitaxel Q3W2/2 (100%)
 Cisplatin-etoposide6/7 (86%)
 Cisplatin-pemetrexed0/3 (0%)
Durvalumab:0.43 (0.15-1.3)0.150.7 (0.2-2.1)0.48
 No n=4120/41 (49%)
 Yes n=65/6 (83%)
Radiation dose:0.99 (0.3-3.4)0.99
 ≤55 Gy n=3217/32 (53%)
 >56 Gy n=158/15 (53%)
Radiation technique:0.8 (0.25-2.5)0.7
 3DCT n=2010/20 (50%)
 VMAT n=2715/27 (56%)
Breathing management:0.66 (0. 3-1.4)0.3
 DIBH n=83/8 (37%)
 FB n=3822/38 (58%)
Adaptive planning due to tumor shrinkage:2.9 (0.7-12)0.131.6 (0.37-6.6)0.53
 Yes n=107/10 (70%)
 No n=3617/36 (47%)
 No-EGFR n=3122/31 (71%)0.35 (0.16-0.7)0.0060.39 (0.18-0.86)0.02
 EGFR n=133/13 (23%)
PDL1 statusb:1 (0.43-2.3)1
 Negative n=116/11 (54%)
 Weak positive n=115/11 (45%)
 Strong positive n=116/11 (54%)
Time interval radiation to surgerya:0.12 (0.02-0.5)0.005
 ≤65 days n=3011/30 (36.7%)
 >65 days n=1714/17 (82%)

Major pathologic regression (MPR), Gray (Gy), Three-dimensional conformal radiation therapy (3DCRT), Volumetric arc therapy (VMAT), Deep inspiratory breath hold (DIBH), Free breathing (FB), Epidermal growth factor receptor (EGFR), Chemoradiation (ChRT).

a

Smoking, histology, and time interval between ChRT and surgery correlated with EGFR mutation (smoking p < 0.0001, histology p = 0.013, time interval between ChRT and surgery p = 0.048). Thus, they were excluded from the MVA.

b

PDL1 status negative (< 1% positive staining); weak positive (1–49%) and strong positive (≥ 50%).

We excluded from the MVA the co-variables that correlated significantly with EGFR: never-smoking (p < 0.0001), adenocarcinoma histology (p = 0.013), and the time interval between radiation and surgery (p = 0.048). Only the no-driver mutation group (OR 0.39, p = 0.02) remained significant for tumor regression on MVA.

Figure 2 compares local control, DFS, and OS at 2 years for the EGFR and no-driver mutation groups. 2-year DFS was lower for the EGFR group: 8% (95% CI 1–29%) vs. 45% (95% CI 29–65%), HR 0.5, p = 0.001. Local control trended lower for the EGFR group: 52% (95% CI 20–76%) vs. 85.8% (95% CI 66.5–95%), HR 0.67, p = 0.17. OS was not significantly different for the EGFR group: 69.2% (95% CI 37–87%) vs. 84% (95% CI 65–93%), HR 1.04, p = 0.86.

(a–c): Kaplan Meier estimated of (a) local control, (b) disease free survival and (c) overall survival for driver mutation (N=16) vs. no driver mutation (N=31).
Figure 2.

(ac): Kaplan Meier estimated of (a) local control, (b) disease free survival and (c) overall survival for driver mutation (N=16) vs. no driver mutation (N=31).

We knew PD-L1 receptor status in 37 cases. Table 4 presents the rates of MPR and pCR and the MRTC between negative, weakly positive, and strongly positive PD-L1 staining. There were no differences in MPR, pCR, or MRTC between the PD-L1 negative, weakly positive or strongly positive subgroups. Figure S2 presents a scatterplot showing MRTC according to the percent of PD-L1 staining, showing no correlation between the percentage of MRTC and the percentage of the PD-L1 positivity (p = 0.5).

Table 4.

Pathologic response to chemoradiation according to and PDL-1 staining

PDL-1 stainingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Negative (N = 13)7/13 (54%)0.91/13 (7.7%)0.5622 (7.2)0.56
Weak positive (N = 13)6/13 (46%)3/13 (23%)29 (7.5)
Strong positive (N = 11)6/11 (54.5%)3/11 (27%)17.3 (7.8)
PDL-1 stainingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Negative (N = 13)7/13 (54%)0.91/13 (7.7%)0.5622 (7.2)0.56
Weak positive (N = 13)6/13 (46%)3/13 (23%)29 (7.5)
Strong positive (N = 11)6/11 (54.5%)3/11 (27%)17.3 (7.8)

Abbreviations: Major pathologic response (MPR), Mean residual tumor cells (MRTC), Pathologic complete response (pCR). PDL-1 negative <1% staining; weak positive 1-49% and strong positive ≥50%.

Table 4.

Pathologic response to chemoradiation according to and PDL-1 staining

PDL-1 stainingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Negative (N = 13)7/13 (54%)0.91/13 (7.7%)0.5622 (7.2)0.56
Weak positive (N = 13)6/13 (46%)3/13 (23%)29 (7.5)
Strong positive (N = 11)6/11 (54.5%)3/11 (27%)17.3 (7.8)
PDL-1 stainingMPR (n, %)p-valuepCR (n, %)p-valueMRTC (%, SD)p-value
Negative (N = 13)7/13 (54%)0.91/13 (7.7%)0.5622 (7.2)0.56
Weak positive (N = 13)6/13 (46%)3/13 (23%)29 (7.5)
Strong positive (N = 11)6/11 (54.5%)3/11 (27%)17.3 (7.8)

Abbreviations: Major pathologic response (MPR), Mean residual tumor cells (MRTC), Pathologic complete response (pCR). PDL-1 negative <1% staining; weak positive 1-49% and strong positive ≥50%.

Discussion

To the best of our knowledge, this is the first study in patients with LA-NSCLC describing pathologic regression as a function of EGFR mutation in surgical specimens following aggressive NA-ChRT using a radiation dose in the neoadjuvant setting similar to the doses used when delivering definitive treatment. We used three pathologic end points; the rate of MPR, the rate of pCR, and the percentage of MRTC to measure response to NA-ChRT. Comparison between the EGFR and the no-driver mutation groups showed lower rates of MPR (23% vs. 71%, p = 0.003) and pCR (0% vs. 26%, p = 0.02) and higher MRTC (43.4% vs. 15.8%, p = 0.009) in the EGFR mutation group. These three parameters all point to the same conclusion, the pathologic regression rates in surgical specimens following NA-ChRT were reduced if an EGFR mutation was present in the tumor.

In contrast to the poor outcomes reported in the EGFR mutation group, the pCR rate of 26% in our no-driver group was consistent with the rate of pCR reported in several large retrospective trials evaluating pathologic response following NA-ChRT in resectable LA-NSCLC. Lococo et al reported a pCR in 22% of cases in a study of 279 patients with LA-NSCLC treated with NA-ChRT,30 and Haque et al, in a large database study, reported the rate of pCR was 17%31 following NA-ChRT. In contrast to these findings, in our study, the rate of pCR in the EGFR group was 0% following an aggressive course of NA-ChRT that used higher RT doses than those used in these studies.

In addition to the poor response in surgical specimens, the DFS of patients with an EGFR mutation was also reduced compared to patients with no-driver mutations (HR 0.5 p = 0.001, Figure 2). Since the NA-ChRT protocol at our institution uses radiation doses commonly used when delivering definitive ChRT without resection, the low rates of pathologic regression and the short duration of DFS in patients with EGFR mutations observed in our study resonates with the poor results observed in patients with EGFR mutations treated with definitive ChRT. A recent study by Butkiewicz et al analyzed the outcome of 205 NSCLC patients treated with definitive platinum-based ChRT as a function of EGFR polymorphism (SNPs). They showed that patients with inoperable disease and functional EGFR genetic variants had poor clinical outcomes. They suggested that EGFR variants associated with elevated gene expression and protein activity may predict the decreased therapeutic efficacy of ChRT and that the EGFR SNPs may act as independent molecular prognostic predictors.32 Akimoto et al showed that the levels of EGFR expression correlated inversely with radiation-induced apoptosis, suggesting that lack of sensitivity to apoptosis induction may be an important mechanism responsible for the radioresistance of tumors with high EGFR expression.33 Chen et al described the role of EGFR in the repair of radiation-induced DNA damage. They suggested that EGFR confers tumor radiation resistance by activating survival and cell proliferation pathways.34 Further studies are required to gain insight into the role of EGFR and other driver mutations in RT-induced cancer cell death.

We found that adenocarcinoma and never-smoking status correlated with reduced tumor regression. These variables are known to be associated with EGFR mutations.35,36 Kayawake et al evaluated the response to ChRT in patients with NSCLC. They showed that patients with adenocarcinoma responded less to ChRT and that only squamous cell carcinoma was associated with achieving a pCR (p = 0.009).3 One of the differences between squamous cell and adenocarcinoma is that the latter is more likely to harbor a driver mutation such as EGFR, ALK, RET, or ROS1 which may explain the findings in Kayawake’s study.37

PD-L1 status did not correlate with pathologic regression measured by MPR, pCR, and MRTC. This finding contrasts with using the PD-L1 biomarker to predict response to immune checkpoint inhibitors in patients with metastatic disease.37,38 Combining treatment with an immune checkpoint inhibitor and radiation is under investigation and may improve response.39 Although durvalumab was used sparingly in this cohort, with only 6 of 31 patients in the no-driver mutation group receiving any durvalumab therapy, cases receiving durvalumab prior to surgery trended towards a higher chance for MPR (83% vs. 53%, OR 0.45, p = 0.16).

Patients with a KRAS mutation had more tumor regression in surgical specimens following ChRT and longer DFS following surgery than patients with an EGFR mutation, yet, trended towards a shorter duration of OS (Figure S1C). We suggest interpreting this finding cautiously due to the small number of patients in our study with KRAS mutations (n = 10). The discrepancy between the favorable rates of pathologic regression observed in our study and the poor prognosis reported for patients with KRAS mutations29,40 deserves further investigation. Enhancing treatment with immunotherapy or downstream inhibitors such as RAF or MEK inhibitors41 may improve clinical outcomes for these patients.

Potential bias in our data may result from selecting a single genetic alteration for analysis without accounting for other mutations that may co-exist and cause undetected interactions. This cohort included several genetic alterations. The rare mutations RET, ROS1, and BRAF were excluded from the EGFR group to produce a homogenous group for UVA and MVA regression analysis. We plan to investigate these rare mutations further in a larger cohort. Also, we had no cases of ALK rearrangement or MET mutation. Although the number of cases in the EGFR and no-driver mutation groups was small, the differences in pathologic response observed between the groups were sufficiently large to be significant. Other biases may result from using durvalumab in six patients within the no-driver mutation group. The improved outcomes with the use of neoadjuvant immunotherapy with chemotherapy compared to chemotherapy alone in patients with Stage IB-IIIA NSCLC reported recently in the Checkmate 816 trial42 suggests that the use of neoadjuvant durvalumab in six patients in the no-driver mutation group may have contributed to the better rates of pathologic regression and longer DFS observed in these patients. However, the proportion of patients who received durvalumab was small and unlikely to have impacted the results of MVA.

TKIs prolong survival significantly in patients with metastatic disease and EGFR-mutated tumors.43 Tyrosine kinase inhibitors were used in our cohort for recurrent disease, thus, explaining the discrepancy between shorter DFS yet similar OS between the driver mutation and the no-driver mutation groups (Figure 2). Since patients received TKIs only after surgery, their use did not affect the rates of pathologic regression observed in the surgical specimens.

For Stage IB-III NSCLC resectable EGFR mutated tumors, the ADUARA trial has shown that adjuvant osimertinib significantly prolonged DFS compared with placebo.12 Treatment with osimertinib has rapidly become the standard of care in these clinical settings. Current studies such as the NEOADAURA trial, which evaluates the use of neoadjuvant osimertinib in patients with resectable NSCLC prior to surgery,13 and the LAURA trial, which is investigating the use of adjuvant osimertinib following ChRT in patients with unresectable LA-NSCLC,14 may further improve outcomes in patients with EGFR mutated tumors treated with ChRT. Current guidelines for patients with unresectable NSCLC suggest treating patients with definitive ChRT and consolidation durvalumab regardless of driver mutation.44 As a result, deferring or not conducting molecular testing reduces the opportunity for patients to participate in clinical trials and potentially derive benefit from early treatment with targeted therapies such as osimertinib.4,5 Given the poor pathologic regression and short duration of DFS in the EGFR cohort detected in our study, we recommend that patients with LA-NSCLC undergo molecular testing for driver mutations allowing for targeted therapies to be introduced earlier in their course of treatment via clinical trials.

Conclusions

Patients with resectable LA-NSCLC and a driver mutation had poor tumor regression in pathology specimens and shorter DFS following NA-ChRT and surgery. PD-L1 was not associated with tumor regression. Future studies in patients with LA-NSCLC should include molecular testing for tumor driver mutations for prognostication and treatment decisions.

Ethics approval

This study was approved by the institutional review board for retrospective analysis (approval number SMC-2678-15).

The authors Sarit Appel, Akram Saad, Jair Bar and Jeffrey Goldstein contributed equally to the work.

Prior presentations

Presented in part at the annual meeting of ESTRO 2021, Madrid, Spain.

REFERENCES

1.

Sequist
LV
,
Yang
JC-H
,
Yamamoto
N
,
O’Byrne
K
,
Hirsh
V
,
Mok
T
, et al. .
Phase III study of Afatinib or cisplatin plus Pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations
.
J Clin Oncol
2013
;
31
:
3327
34
. doi: https://doi.org/10.1200/JCO.2012.44.2806

2.

Mok
TS
,
Wu
Y-L
,
Thongprasert
S
,
Yang
C-H
,
Chu
D-T
,
Saijo
N
, et al. .
Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma
.
N Engl J Med
2009
;
361
:
947
57
. doi: https://doi.org/10.1056/NEJMoa0810699

3.

Kayawake
H
,
Okumura
N
,
Yamanashi
K
,
Takahashi
A
,
Itasaka
S
,
Yoshioka
H
, et al. .
Non-small cell lung cancer with pathological complete response: predictive factors and surgical outcomes
.
Gen Thorac Cardiovasc Surg
2019
;
67
:
773
81
. doi: https://doi.org/10.1007/s11748-019-01076-9

4.

Gross
CP
,
Meyer
CS
,
Ogale
S
,
Kent
M
,
Wong
WB
.
Associations between Medicaid insurance, biomarker testing, and outcomes in patients with advanced NSCLC
.
J Natl Compr Canc Netw
2022
;
20
:
479
487
. doi: https://doi.org/10.6004/jnccn.2021.7083

5.

Roberts
TJ
,
Kesselheim
AS
,
Avorn
J
.
Variation in use of lung cancer targeted therapies across state Medicaid programs, 2020-2021
.
JAMA Netw Open
2023
;
6
( ): e2252562 . doi: https://doi.org/10.1001/jamanetworkopen.2022.52562

6.

Faivre-Finn
C
,
Vicente
D
,
Kurata
T
,
Planchard
D
,
Paz-Ares
L
,
Vansteenkiste
JF
, et al. .
Four-year survival with Durvalumab after Chemoradiotherapy in stage III NSCLC-an update from the PACIFIC trial
.
J Thorac Oncol
2021
;
16
:
860
67
. doi: https://doi.org/10.1016/j.jtho.2020.12.015

7.

Antonia
SJ
,
Villegas
A
,
Daniel
D
,
Vicente
D
,
Murakami
S
,
Hui
R
, et al. .
Durvalumab after Chemoradiotherapy in stage III non-small-cell lung cancer
.
N Engl J Med
2017
;
377
:
1919
29
. doi: https://doi.org/10.1056/NEJMoa1709937

8.

Bradley
JD
,
Hu
C
,
Komaki
RR
,
Masters
GA
,
Blumenschein
GR
,
Schild
SE
, et al. .
Long-term results of NRG oncology RTOG 0617: Standard- versus high-dose Chemoradiotherapy with or without Cetuximab for Unresectable stage III non-small-cell lung cancer
.
J Clin Oncol
2020
;
38
:
706
14
. doi: https://doi.org/10.1200/JCO.19.01162

9.

Wu
R
,
Yu
S
,
Ye
J
,
Wang
Y
,
Zhao
Z
,
Liu
H
, et al. .
A multicenter retrospective study on the prognosis of stage III Unresectable mutant non-small cell lung cancer with tyrosine kinase inhibitors therapy
.
Front Oncol
2021
;
11
: 692703 . doi: https://doi.org/10.3389/fonc.2021.692703

10.

Ready
N
,
Jänne
PA
,
Bogart
J
,
Dipetrillo
T
,
Garst
J
,
Graziano
S
, et al. .
Chemoradiotherapy and Gefitinib in stage III non-small cell lung cancer with Epidermal growth factor receptor and KRAS Mutation analysis: cancer and leukemia group B (CALEB) 30106, a CALGB-stratified phase II trial
.
J Thorac Oncol
2010
;
5
:
1382
90
. doi: https://doi.org/10.1097/JTO.0b013e3181eba657

11.

Wu
Y-L
,
Tsuboi
M
,
He
J
,
John
T
,
Grohe
C
,
Majem
M
, et al. .
Osimertinib in Resected EGFR-Mutated non-small-cell lung cancer
.
N Engl J Med
2020
;
383
:
1711
23
. doi: https://doi.org/10.1056/NEJMoa2027071

12.

Herbst
RS
,
Wu
Y-L
,
John
T
,
Grohe
C
,
Majem
M
,
Wang
J
, et al. .
Adjuvant Osimertinib for Resected EGFR-Mutated stage IB-IIIA non-small-cell lung cancer: updated results from the phase III randomized ADAURA trial
.
JCO
2023
;
41
:
1830
40
. doi: https://doi.org/10.1200/JCO.22.02186

13.

Tsuboi
M
,
Weder
W
,
Escriu
C
,
Blakely
C
,
He
J
,
Dacic
S
, et al. .
Neoadjuvant Osimertinib with/without chemotherapy versus chemotherapy alone for EGFR-Mutated Resectable non-small-cell lung cancer: Neoadaura
.
Future Oncol
2021
;
17
:
4045
55
. doi: https://doi.org/10.2217/fon-2021-0549

14.

Lu
S
,
Casarini
I
,
Kato
T
,
Cobo
M
,
Özgüroğlu
M
,
Hodge
R
, et al. .
Osimertinib maintenance after definitive Chemoradiation in patients with Unresectable EGFR Mutation positive stage III non-small-cell lung cancer: LAURA trial in progress
.
Clin Lung Cancer
2021
;
22
:
371
75
. doi: https://doi.org/10.1016/j.cllc.2020.11.004

15.

Zens
P
,
Bello
C
,
Scherz
A
,
Koenigsdorf
J
,
Pöllinger
A
,
Schmid
RA
, et al. .
A Prognostic score for non-small cell lung cancer Resected after Neoadjuvant therapy in comparison with the tumor-node-metastases classification and major pathological response
.
Mod Pathol
2021
;
34
:
1333
44
. doi: https://doi.org/10.1038/s41379-021-00777-y

16.

Weissferdt
A
,
Pataer
A
,
Vaporciyan
AA
,
Correa
AM
,
Sepesi
B
,
Moran
CA
, et al. .
Agreement on major pathological response in NSCLC patients receiving Neoadjuvant chemotherapy
.
Clin Lung Cancer
2020
;
21
:
341
48
. doi: https://doi.org/10.1016/j.cllc.2019.11.003

17.

Machtay
M
,
Lee
JH
,
Stevenson
JP
,
Shrager
JB
,
Algazy
KM
,
Treat
J
, et al. .
Two commonly used Neoadjuvant Chemoradiotherapy regimens for locally advanced stage III non-small cell lung carcinoma: long-term results and associations with pathologic response
.
J Thorac Cardiovasc Surg
2004
;
127
:
108
13
. doi: https://doi.org/10.1016/j.jtcvs.2003.07.027

18.

Sonett
JR
,
Suntharalingam
M
,
Edelman
MJ
,
Patel
AB
,
Gamliel
Z
,
Doyle
A
, et al. .
Pulmonary resection after curative intent radiotherapy (>59 GY) and concurrent chemotherapy in non-small-cell lung cancer
.
Ann Thorac Surg
2004
;
78
:
1200
1205
. doi: https://doi.org/10.1016/j.athoracsur.2004.04.085

19.

Friedel
G
,
Budach
W
,
Dippon
J
,
Spengler
W
,
Eschmann
SM
,
Pfannenberg
C
, et al. .
Phase II trial of a Trimodality regimen for stage III non-small-cell lung cancer using chemotherapy as induction treatment with concurrent Hyperfractionated Chemoradiation with carboplatin and paclitaxel followed by subsequent resection: a single-center study
.
J Clin Oncol
2010
;
28
:
942
48
. doi: https://doi.org/10.1200/JCO.2008.21.7810

20.

Hellmann
MD
,
Chaft
JE
,
William
WN
,
Rusch
V
,
Pisters
KMW
,
Kalhor
N
, et al. .
Pathological response after Neoadjuvant chemotherapy in Resectable non-small-cell lung cancers: proposal for the use of major pathological response as a Surrogate Endpoint
.
Lancet Oncol
2014
;
15
:
e42
50
. doi: https://doi.org/10.1016/S1470-2045(13)70334-6

21.

Junker
K
,
Langner
K
,
Klinke
F
,
Bosse
U
,
Thomas
M
.
Grading of tumor regression in non-small cell lung cancer: morphology and prognosis
.
Chest
2001
;
120
:
1584
91
. doi: https://doi.org/10.1378/chest.120.5.1584

22.

Travis
WD
,
Dacic
S
,
Wistuba
I
,
Sholl
L
,
Adusumilli
P
,
Bubendorf
L
, et al. .
IASLC Multidisciplinary recommendations for pathologic assessment of lung cancer resection specimens after Neoadjuvant therapy
.
J Thorac Oncol
2020
;
15
:
709
40
. doi: https://doi.org/10.1016/j.jtho.2020.01.005

23.

Pirker
R
,
Herth
FJF
,
Kerr
KM
,
Filipits
M
,
Taron
M
,
Gandara
D
, et al. .
Consensus for EGFR Mutation testing in non-small cell lung cancer: results from a European workshop
.
J Thorac Oncol
2010
;
5
:
1706
13
. doi: https://doi.org/10.1097/JTO.0b013e3181f1c8de

24.

Xue
Y
,
R. Wilcox
W
.
Changing paradigm of cancer therapy: precision medicine by next-generation sequencing
.
Cancer Biol Med
2016
;
13
:
12
18
. doi: https://doi.org/10.20892/j.issn.2095-3941.2016.0003

25.

Kruglyak
KM
,
Lin
E
,
Ong
FS
.
Next-generation sequencing and applications to the diagnosis and treatment of lung cancer
.
Adv Exp Med Biol
2016
;
890
:
123
36
. doi: https://doi.org/10.1007/978-3-319-24932-2_7

26.

Takada
K
,
Okamoto
T
,
Toyokawa
G
,
Kozuma
Y
,
Matsubara
T
,
Haratake
N
, et al. .
The expression of PD-L1 protein as a Prognostic factor in lung squamous cell carcinoma
.
Lung Cancer
2017
;
104
:
7
15
. doi: https://doi.org/10.1016/j.lungcan.2016.12.006

27.

Appel
S
,
Goldstein
J
,
Perelman
M
,
Rabin
T
,
Urban
D
,
Onn
A
, et al. .
Neo-adjuvant Chemo-radiation to 60 gray followed by surgery for locally advanced non-small cell lung cancer patients: evaluation of Trimodality strategy
.
Isr Med Assoc J
2017
;
19
:
614
19
.

28.

Appel
S
,
Bar
J
,
Ben-Nun
A
,
Perelman
M
,
Alezra
D
,
Urban
D
, et al. .
Comparative effectiveness of intensity modulated radiation therapy to 3-dimensional Conformal radiation in locally advanced lung cancer: pathological and clinical outcomes
.
Br J Radiol
2019
;
92
:
20180960
: 20180960 . doi: https://doi.org/10.1259/bjr.20180960

29.

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: https://doi.org/10.1016/j.jtho.2019.01.020

30.

Lococo
F
,
Sassorossi
C
,
Nachira
D
,
Chiappetta
M
,
Petracca Ciavarella
L
,
Vita
E
, et al. .
Prognostic factors and long-term survival in locally advanced NSCLC with pathological complete response after surgical resection following Neoadjuvant therapy
.
Cancers (Basel)
2020
;
12
( ): 3572 . doi: https://doi.org/10.3390/cancers12123572

31.

Haque
W
,
Verma
V
,
Butler
EB
,
Teh
BS
.
Pathologic nodal clearance and complete response following Neoadjuvant Chemoradiation for clinical N2 non-small cell lung cancer: predictors and long-term outcomes
.
Lung Cancer
2019
;
130
:
93
100
. doi: https://doi.org/10.1016/j.lungcan.2019.02.003

32.

Butkiewicz
D
,
Krześniak
M
,
Gdowicz-Kłosok
A
,
Giglok
M
,
Marszałek-Zeńczak
M
,
Suwiński
R
.
Polymorphisms in EGFR gene predict clinical outcome in Unresectable non-small cell lung cancer treated with radiotherapy and platinum-based Chemoradiotherapy
.
Int J Mol Sci
2021
;
22
( ): 5605 . doi: https://doi.org/10.3390/ijms22115605

33.

Akimoto
T
,
Hunter
NR
,
Buchmiller
L
,
Mason
K
,
Ang
KK
,
Milas
L
.
Inverse relationship between Epidermal growth factor receptor expression and Radiocurability of murine Carcinomas
.
Clin Cancer Res
1999
;
5
:
2884
90
.

34.

Chen
DJ
,
Nirodi
CS
.
The Epidermal growth factor receptor: a role in repair of radiation-induced DNA damage
.
Clin Cancer Res
2007
;
13
:
6555
60
. doi: https://doi.org/10.1158/1078-0432.CCR-07-1610

35.

Viñolas
N
,
Garrido
P
,
Isla
D
,
Provencio
M
,
Majem
M
,
Artal
A
, et al. .
Lung cancer in never-smoking women: A sub-analysis of the Spanish female-specific database World07
.
Cancer Invest
2017
;
35
:
358
65
. doi: https://doi.org/10.1080/07357907.2017.1295461

36.

Yano
T
,
Haro
A
,
Shikada
Y
,
Maruyama
R
,
Maehara
Y
.
“Non-small cell lung cancer in never Smokers as a representative 'non-smoking-associated lung cancer': epidemiology and clinical features”
.
Int J Clin Oncol
2011
;
16
:
287
93
. doi: https://doi.org/10.1007/s10147-010-0160-8

37.

Shea
M
,
Costa
DB
,
Rangachari
D
.
Management of advanced non-small cell lung cancers with known mutations or Rearrangements: latest evidence and treatment approaches
.
Ther Adv Respir Dis
2016
;
10
:
113
29
. doi: https://doi.org/10.1177/1753465815617871

38.

Shi
Y
,
Duan
J
,
Guan
Q
,
Xue
P
,
Zheng
Y
.
Effectivity and safety of PD-1/PD-L1 inhibitors for different level of PD-L1-positive, advanced NSCLC: A meta-analysis of 4939 patients from randomized controlled trials
.
Int Immunopharmacol
2020
;
84
:
106452
. doi: https://doi.org/10.1016/j.intimp.2020.106452

39.

Geng
Y
,
Zhang
Q
,
Feng
S
,
Li
C
,
Wang
L
,
Zhao
X
, et al. .
Safety and efficacy of PD-1/PD-L1 inhibitors combined with radiotherapy in patients with non-small-cell lung cancer: a systematic review and meta-analysis
.
Cancer Med
2021
;
10
:
1222
39
. doi: https://doi.org/10.1002/cam4.3718

40.

Ferrer
I
,
Zugazagoitia
J
,
Herbertz
S
,
John
W
,
Paz-Ares
L
,
Schmid-Bindert
G
.
KRAS-mutant non-small cell lung cancer: from biology to therapy
.
Lung Cancer
2018
;
124
:
53
64
. doi: https://doi.org/10.1016/j.lungcan.2018.07.013

41.

Uras
IZ
,
Moll
HP
,
Casanova
E
.
Targeting KRAS mutant non-small-cell lung cancer: past, present and future
.
Int J Mol Sci
2020
;
21
( ): 4325 . doi: https://doi.org/10.3390/ijms21124325

42.

Forde
PM
,
Spicer
J
,
Girard
N
.
Neoadjuvant Nivolumab plus chemotherapy in Resectable lung cancer
.
N Engl J Med
2022
;
387
:
572
73
. doi: https://doi.org/10.1056/NEJMc2208133

43.

Soria
J-C
,
Ohe
Y
,
Vansteenkiste
J
,
Reungwetwattana
T
,
Chewaskulyong
B
,
Lee
KH
, et al. .
Osimertinib in untreated EGFR-Mutated advanced non-small-cell lung cancer
.
N Engl J Med
2018
;
378
:
113
25
. doi: https://doi.org/10.1056/NEJMoa1713137

44.

National Comprehensive Cancer Network
.
2023
.
Available from
: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 Unported License http://creativecommons.org/licenses/by-nc/4.0/, which permits unrestricted non-commercial reuse, provided the original author and source are credited.

Supplementary data