Evidence from resected early-stage non-small cell lung cancer with EGFR mutation: a literature review
Introduction
The presence of sensitizing mutation in the epidermal growth factor receptor (EGFR) gene is a major driver mutation in non-small cell lung cancer (NSCLC), with specific clinical and therapeutic implications (1).
In advanced-stage disease, EGFR mutations are present in 20–22% of NSCLC patients in Western countries and India and in 30% of patients in Japan while the rates observed in East Asia rise to 47–64% (2-4). First line targeted therapy with first generation EGFR-tyrosine-kinase inhibitors (TKIs) such as gefitinib and erlotinib showed better outcomes than chemotherapy (5,6). Subsequently, newer generation EGFR-TKIs such us afatinib and dacomitinib (second generation) and osimertinib (third generation) have further improved progression-free survival (PFS) compared with first-generation TKIs, with osimertinib demonstrating an overall survival (OS) advantage and a more favourable safety profile (7-10).
In early-stage NSCLC [i.e., stage I to resectable IIIA tumor-node-metastasis (TNM) 8th American Joint Committee on Cancer (AJCC) edition], surgery and/or radiotherapy are usually performed with curative intent, with adjuvant or neoadjuvant chemotherapy being added in some cases to improve outcomes, regardless of molecular biomarker status. Since the development of targeted therapy in advanced-stage NSCLC, there has been increasing interest in the epidemiology, characteristics and therapeutic implications of EGFR mutation in early-stage NSCLC.
The aim of this review is to summarize the available evidence in early-stage EGFR mutant NSCLC, including epidemiological issues, clinical, radiological and pathological characteristics; and therapeutic implications of EGFR-TKIs in the adjuvant and neoadjuvant settings. We present this article in accordance with the Narrative Review reporting checklist (available at https://cco.amegroups.com/article/view/10.21037/cco-24-35/rc).
Methods
We conducted a comprehensive review of the literature for information on NSCLC patients with either EGFR mutant or in early-stage NSCLC, including evidence published before 31st December 2023.
Sources consulted included major international databases such as MEDLINE, PubMed, Scopus and Web of Science; as well as abstracts presented at major international lung cancer congresses such as the American Society of Clinical Oncology (ASCO), the European Society of Medical Oncology (ESMO) and the World Conference on Lung Cancer (WCLC) of the International Association for the Study of Lung Cancer (IASLC).
Information included was published in English and availability of full text (for papers) or full presentation (for congress communications) was required. Clinical trials, systematic reviews and observational studies were included, whereas clinical case reports and narrative reviews were excluded.
The search strategy is summarized in Table 1.
Table 1
Items | Specification |
---|---|
Date of search | January 3rd 2024 |
Databases and other sources searched | PubMed, MEDLINE, Scopus, Web of Science, ASCO, ESMO and WCLC annual congresses |
Search terms used | [“Non-Small Cell Lung Cancer” OR NSCLC] AND [“EGFR mutant”] OR [“Early stage” OR Resectable] |
Timeframe | Evidence published before 31st December 2023 |
Inclusion and exclusion criteria | Inclusion criteria: |
- Clinical trials, observational studies, systematic reviews, meta-analysis | |
- Written in English | |
- Full text (journal papers) or full presentation (congress communications) available | |
Exclusion criteria: | |
- Narrative reviews, opinion articles or case report studies | |
- Written in other languages different than English | |
- No full text available | |
Selection process | Abstracts were reviewed by the authors and considered for the summary if fit within the scope of the review |
ASCO, American Society of Clinical Oncology; ESMO, European Society of Medical Oncology; WCLC, World Conference on Lung Cancer; NSCLC, non-small cell lung cancer; EGFR, epidermal growth factor receptor.
Diagnosis and epidemiology: clinical, radiological, pathological and molecular features
EGFR mutations are the most common driver mutations found in NSCLC. The most common variants are the exon 19 deletion and the L858R exon 21 mutation, being known as classical or sensitizing mutations (1). Other less common mutations include G719X, S768I and L861Q between exons 18 and 21 and exon 20 insertions (1,11). All these mutations cause a constitutive activation of the tyrosine kinase domain in the EGFR receptor, leading to uncontrolled cell growth; however, the sensitivity to TKI therapy differs, being greater in the classical mutations than in the uncommon ones. Genetic sequencing of EGFR exons 18 to 21 is required to diagnose these mutations, as this improves the sensitivity of mutation-specific based polymerase chain reaction (PCR) (12).
NSCLC is diagnosed at an early-stages (stage I to IIIA of the 8th AJCC TNM edition) in approximately 30% of NSCLC cases (13). EGFR mutation rates vary between geographical regions, ranging from 13–17% in Western countries (14,15) to 53–58% in East Asian countries (16-18). Regardless of the geographic region, EGFR mutation rates appear to be slightly higher in early-stages than in advanced-stages, being higher in stage IA (14,15,17,18).
In terms of clinical characteristics, EGFR mutation is more common in women and in never-smokers than in men and smokers, like it is in advanced-stages (14-18). Radiologically, EGFR mutant early-stage NSCLC often presents with smaller solid consolidations and the presence of ground-glass opacities (17), with lower glucose uptake levels on positron emission tomography (PET) scan (18). In terms of histological pattern, EGFR mutations are more common in acinar or papillary pattern low-grade adenocarcinomas, and less common in tumors with sarcomatoid features, pleomorphic differentiation or in adenosquamous carcinomas (17,18).
At the molecular level, classic sensitizing mutations reach close to 90% EGFR mutations in early-stages (both exon 19 deletions and L858R exon 21 mutation); L858R mutations show a higher proportion than the observed in advanced-stages (15,17-20). TP53 co-mutation, which is a negative prognostic feature, is less common in early-stage than in advanced-stage disease (27% versus 65%) (15). Among the uncommon mutations, exon 20 insertions are less frequent than in advanced disease (15).
According to patient cohorts treated without adjuvant targeted therapy (before adjuvant EGFR-TKIs were evaluated), the prognosis in low risk resected EGFR mutant early-stage NSCLC is similar to non-mutant early-stage NSCLC: recurrence-free survival (RFS) rates and median RFS are close (14,17,18). However, according to the control arms of recent clinical trials, a higher risk of recurrence is suggested particularly in stage II–IIIA and 5 years after surgical treatment (18,21,22). Local recurrence and systemic recurrence rates are similar in EGFR mutant and non-mutant NSCLC, although brain metastases at recurrence are more common in EGFR mutant patients (9–16% versus 6–12%) (17-20). OS may be better in EGFR mutant early-stage NSCLC due to the availability of targeted therapy in the advanced-stage setting, in the event of progression, which improves outcomes in cases with distant recurrence (14,17,18).
Local treatment, adjuvant chemotherapy and immunotherapy
Regardless of the presence of an EGFR mutation, the standard local treatment of early-stage NSCLC requires complete tumor resection with an anatomical resection (mainly lobectomy) with systematic mediastinal lymph node dissection (23,24).
Clinical trials evaluating sub lobar resection for small and peripheral tumors have not reported subgroup results according to the presence or absence of driver mutations (25,26). However, the benefit seen in the overall population results may translate to the same benefit in EGFR mutant population given the similar local and locoregional recurrence rates observed (17,18).
In all commers, after resection, adjuvant chemotherapy with four cycles of cisplatin-based doublet is recommended after resection for resected high-risk stage IB (mostly tumors larger than 4 cm), stage II and stage III (7th AJCC TNM edition) NSCLC, achieving a 5% OS improvement at 5 years after surgery, with a number needed to treat (NNT) of 20–25 (27-30). This means that 20 to 25 patients should be treated with chemotherapy to prevent one death. The role of neoadjuvant chemotherapy (i.e., before surgery) shows similar OS results (31,32).
Recently, the combination of immunotherapy with anti-PD-(L)1 immune checkpoint inhibitors with chemotherapy has improved outcomes in both the adjuvant and the neoadjuvant settings. However, immunotherapy has not been shown to be beneficial in advanced EGFR-mutant NSCLC (33-37). Therefore, the presence of an EGFR mutation is often an exclusion criterion (or at least a stratification factor) in the major clinical trials of immunotherapy in early-stage disease, and major regulatory agencies exclude EGFR mutated [and anaplastic leukemia kinase (ALK), altered] NSCLC in the approval of these treatments. Adjuvant immunotherapy with atezolizumab improved outcomes in the overall population in the Impower010 clinical trial, but did not appear to improve outcomes in the EGFR-mutant subset of patients; similarly, the PEARLS clinical trial of adjuvant therapy with pembrolizumab did not include enough EGFR mutant cases to draw conclusions (22,38). In the neoadjuvant or perioperative setting, the presence of an EGFR mutation was an exclusion criterion in many trials [NADIM, NADIM2, Checkmate-816, Checkmate-77T and AEGEAN (39-43)], while in others its detection and reporting was not mandatory and the number of patients with mutation status reported was very limited to draw conclusions [Keynote-671 (44)].
Therefore, most clinical practice guidelines do not recommend the use of immunotherapy in the treatment of patients with early-stage EGFR mutant NSCLC (30,45,46).
Targeted therapy in the adjuvant setting
The treatment benefit of EGFR-TKIs in advanced disease has resulted in the study of their role in early stages (5-9).
First generation EGFR-TKI
The main clinical trials investigating adjuvant treatment with first generation inhibitors are summarized in Table 2. These trials included only patients with classic sensitizing EGFR mutations and their inclusion criteria were based on the 7th edition of the AJCC TNM.
Table 2
Clinical trial | Country (year) | Phase | Patient number | Stage | Treatment | RFS; HR (95% CI) | OS; HR (95% CI) | Received EGFR-TKI at relapse |
---|---|---|---|---|---|---|---|---|
RADIANT* (47) (NCT00373425) | International (2007–2010) | III | 161 | IB–IIIA | After CT: erlotinib 2 y vs. placebo | 46.4 vs. 28.5 m; 0.61 (0.38–0.98) | NR vs. NR; 1.09 (0.52–2.16) | Not reported |
ADJUVANT-CTONG1104 (48) (NCT01405079) | China (2011–2014) | III | 222 | II–IIIA | Gefitinib 2 y vs. CT | 30.8 vs. 19.8 m; 0.56 ( 0.40–0.79) | 75.5 vs. 62.8 m; 0.92 (0.62–1.36) | 52% |
IMPACT (49) (UMIN000006252) | China-Japan (2011–2015) | III | 234 | II–IIIA | Gefitinib 2 y vs. CT | 35.9 vs. 25.1 m; 0.92 (0.67–1.28) | NR vs. NR; 1.03 (0.65–1.65) | 52% |
EVAN (50) (NCT01683175) | China (2012–2015) | II | 102 | IIIA | Erlotinib 2 y vs. CT | 42.4 vs. 21.0 m; 0.27 (0.14–0.53) | 84.2 vs. 61.1 m; 0.32 (0.15–0.67) | 37% |
EVIDENCE (51) (NCT02448797) | China (2015–2019) | III | 322 | II–IIIA | Icotinib 2 y vs. CT | 47.0 vs. 22.1 m; 0.36 (0.24–0.55) | NR vs. NR; 0.91 (0.42–1.94) | Not reported |
CORIN (52) (NCT02264210) | China (2015–2020) | III | 128 | IB | Icotinib 1 y vs. placebo | NR vs. NR; 0.23 (0.07–0.81) | NR vs. NR; NR | 83% |
*, only patients with EGFR mutant NSCLC. EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitors; NSCLC, non-small cell lung cancer; RFS, recurrence-free survival; OS, overall survival; CT, chemotherapy; y, years; m, months; HR, hazard ratio; CI, confidence interval; NR, not reached.
The role of adjuvant erlotinib was evaluated in the RADIANT trial, which compared 2 years of erlotinib versus placebo after complete resection and adjuvant chemotherapy (47). Although a benefit in RFS was demonstrated, this did not translate into a benefit in OS. Likewise, the role of adjuvant gefitinib was evaluated in the ADJUVANT and IMPACT, both compared to chemotherapy (48,49). In these trials, a benefit in RFS was observed but again there were no differences in OS. The lack of OS benefit in first generation TKI trials may be due to therapeutic options at relapse, as 55–83% patients in these trials received an EGFR-TKI at disease recurrence.
The lack of benefit in OS with adjuvant first generation TKIs was confirmed in two meta-analyses that included data from these previously mentioned trials and several single-arm earlier phase studies (53,54).
Subsequently, two clinical trials investigated the role of icotinib, another first generation TKI. The EVIDENCE trial enrolled patients with stage II to IIIA disease and compared icotinib with chemotherapy, while the CORIN trial enrolled patients with stage IB disease and compared icotinib with placebo (51,52). Both trials observed an RFS benefit but no OS differences, although OS data may still not be mature yet, as the phase 2 ICOMPARE trial in stage II to IIIA patients treated with icotinib showed an RFS and OS benefit with 2 years of adjuvant treatment compared to 1 year of treatment (55).
Finally, the EVAN trial, which included patients with resected stage IIIA who were treated with erlotinib versus placebo for 2 years in China, observed both an RFS and an OS benefit in the experimental arm (50). However, adjuvant treatment may would have had a greater effect due to the inclusion of higher-risk patients, the lack of chemotherapy in the control arm and the low rate of EGFR-TKI treatment at progression (32%, lower than in the previous trials).
Third generation EGFR-TKI
The ADAURA clinical trial evaluated the adjuvant therapy with third generation TKI osimertinib (21,56-59). Previously, osimertinib had demonstrated an advantage over first-generation TKIs in terms of efficacy [including in central nervous system (CNS) disease] and toxicity in advanced disease (8,10,60).
ADAURA was a randomized phase 3 clinical trial that enrolled 682 patients with Eastern Cooperative Oncology Group (ECOG) performance status 0 to 1 with a classic sensitizing EGFR mutation (19 exon deletion or 21 exon L858R mutation) in stages IB to IIIA (7th AJCC TNM edition), after complete resection with or without adjuvant chemotherapy. Patients were randomized 1:1 to receive osimertinib or placebo for 3 years. The primary endpoint was RFS in patients with stage II to IIIA disease. In the study population 64% of patients were from East Asia, 64% were women and 70% were never smokers. The stage distribution was 32%, 34% and 34% for IB, II and IIIA respectively. Prior to trial treatment, 60% of patients received chemotherapy at investigator’s decision. All patients were required to have a basal brain imaging study prior to enrolment.
Table 3 shows the main efficacy results of ADAURA study.
Table 3
Outcome | Osimertinib (95% CI) | Placebo (95% CI) | HR (95% CI) |
---|---|---|---|
DFS stages II–IIIA | |||
Median DFS, months | 65.8 (54.4–NR) | 21.9 (16.6–27.5) | |
DFS at 24 months | 90% (84–93%) | 44% (37–51%) | 0.23 (0.18–0.30) |
DFS at 36 months | 84% (unknown) | 34% (unknown) | |
DFS at 48 months | 70% (62–76%) | 29% (23–35%) | |
DFS stages IB–IIIA | |||
Median DFS, months | 65.8 (61.7–NR) | 28.1 (22.1–35) | |
DFS at 24 months | 89% (85–92%) | 52% (46–58%) | 0.27 (0.21–0.34) |
DFS at 36 months | 85% (unknown) | 44% (unknown) | |
DFS at 48 months | 73% (67–78%) | 38% (32–43%) | |
Intracranial DFS (DFS CNS at 48 months) | |||
Stages II–IIIA | 90% (85–94%) | 75% (67–81%) | 0.24 (0.14–0.42) |
Stages IB–IIIA | 92% (88–95%) | 81% (75–85%) | 0.36 (0.23–0.57) |
OS stages IB–IIIA | |||
Median OS, months | NR (NR–NR) | NR (NR–NR) | |
OS at 36 months | 95% (unknown) | 89% (unknown) | 0.49 (0.34–0.70) |
OS at 48 months | 93% (unknown) | 84% (unknown) | |
OS at 60 months | 88% (83–91%) | 78% (73–82%) |
CI, confidence interval; HR, hazard ratio; DFS, disease-free survival; NR, not reached; CNS, central nervous system; OS, overall survival.
Osimertinib treatment significantly improved RFS compared to placebo in stage II to IIIA (primary endpoint) and in the overall population (stage IB to IIIA, secondary endpoint). In subgroup analysis, the benefit was maintained regardless of age, gender, race, smoking history, stage, EGFR mutation and chemotherapy administration (21).
In terms of the pattern of relapse, both local and distant recurrences were lower in patients treated with osimertinib, with fewer recurrences in the most common sites (lung, CNS, lymph nodes and bone). Specifically, for CNS recurrence, osimertinib treatment showed an absolute risk reduction of 11% at 4 years follow up [from 92 with placebo to 81% with osimertinib, hazard ratio (HR) 0.36] (56).
The final OS analysis was then reported and showed a benefit with osimertinib treatment with an absolute risk reduction of 10% for death at 5 years (88% with placebo versus 78% with osimertinib, HR 0.49). The OS benefit was consistent in all pre-specified subgroups, including stage (although the magnitude of the benefit appeared to be greater in stage IIIA than in IB) and chemotherapy use (59).
Osimertinib treatment also prolonged time to subsequent oncological treatments (59). Among placebo-treated patients who received subsequent treatment, 88% of them received an EGFR-TKI: this proves that the OS benefit is not due to the lack of active treatment at relapse. The most common TKI was osimertinib (43%), followed by first generation TKIs (gefitinib 30%, erlotinib 13%, icotinib 8%), second generation afatinib (16%) and dacomitinib (1%) as well as other third generation TKI (aumolertinib 2%, furmonertinib 1%).
Osimertinib-related toxicity was 91% of any grade and 23% of grade 3 or higher (21,56). The toxicity profile was similar to that seen in advanced disease, with diarrhoea (47%), paronychia (27%) and dry skin (25%). Interstitial lung disease and cardiac events occurred in 3% and 6% of patients, respectively. Dose reduction, interruption and discontinuation rates were 12%, 27% and 13%, respectively: dose reduction and interruption rates were higher than those reported in advanced disease (while discontinuation rates were similar), suggesting differences in disease and toxicity perception and in reporting between different disease settings (8). Despite this, osimertinib did not adversely affect quality of life compared with placebo (57).
Based on these results, osimertinib has been approved by the major regulatory authorities in the US, Europa and China [Food and Drug Administration (FDA), European Medicines Agency (EMA) and National Medical Products Administration (NMPA)], among others, and its use is recommended as adjuvant treatment in patients with IB–IIIA NSCLC after complete resection with or without adjuvant chemotherapy (30,45,46). This is the first approval and recommendation of a targeted therapy treatment in early-stage NSCLC, which makes it necessary to test for EGFR mutations in these stages (as do the recommendation against prescribing adjuvant or neoadjuvant immunotherapy in the presence of an EGFR mutation).
Following the recent publication of these findings, some concerns have been raised about adjuvant treatment with osimertinib.
Firstly, the optimal duration of the treatment is unclear. Kaplan-Meier RFS curves appear to show an increase in recurrence in the osimertinib arm after 3 years (the duration of treatment) (21,56). In addition, EGFR mutation is associated with a later pattern of relapse (18). It has been suggested that a longer adjuvant treatment with osimertinib may improve the efficacy outcomes of this treatment. Therefore, the TARGET trial (NCT 05526755) is ongoing, evaluating 5 years of adjuvant treatment with osimertinib. Further follow-up in the ADAURA trial may provide more information on this issue.
Secondly, the role of adjuvant chemotherapy is unclear. In the ADAURA trial, subgroup analyses suggest that the benefit of osimertinib is independent of the use of chemotherapy (21,56,58). The use of chemotherapy was at the discretion of the investigator and it was more common in patients younger than 70 years and with stage II to IIIA disease (58). While this supports the use of adjuvant osimertinib in patients who are not fit enough to receive chemotherapy or in tumors for which there is no indication of adjuvant chemotherapy, it does not mean that chemotherapy can be safely omitted in other subgroups and so it is still recommended according to tumor stage and patient characteristics. Recent clinical trials also suggest a benefit in combining chemotherapy with osimertinib in advanced disease (61,62), but whether if this benefit translates to adjuvant therapy (and in which patients) is still unclear.
Thirdly, prognostic and predictive biomarkers are needed. Since in the ADAURA trial 30% of patients in the osimertinib arm had a recurrence at 4 years, risk stratification may select candidates for treatment intensification (56). Thus, persistence of circulating tumoral DNA (ctDNA) or minimal residual disease (MRD) after surgery, the presence of concomitant mutations in TP53 or RB1 genes and the presence of PD-L1 expression have been suggested as negative prognostic factors (63-66).
Fourth and finally, several settings are not assessed in the ADAURA trial, such as tumors smaller than 4 cm, the presence of affected surgical margins or tumors treated with ablative radiotherapy. Some studies are evaluating the role of osimertinib after complete resection in stage IA2–IA3 EGFR NSCLC, where the frequency of EGFR mutation is higher (ADAURA2, NCT-05120349), or after radical chemoradiotherapy in unresectable locally advanced NSCLC (LAURA, NCT-03521154). Patients with positive surgical margins who require adjuvant radiotherapy and patients with incidental stage IIIB–IIIC after complete resection are likely to benefit from adjuvant treatment with osimertinib, although this was not represented in the clinical trial (21).
Other clinical trials investigation adjuvant treatment with third generation TKIs are ongoing, including trials comparing furmonertinib with placebo (NCT-04853342, FORWARD trial); aumolertinib with placebo (NCT-04687241) or aumolertinib with chemotherapy versus aumolertinib alone versus chemotherapy alone (NCT-04762459, APEX trial).
Targeted therapy in the neoadjuvant setting
The evidence for EGFR-TKIs as neoadjuvant therapy is limited, so their use is not recommended in routine clinical practice and is restricted to the investigational setting.
With regard to first generation TKIs, the EMERGING clinical trial (CTONG-1103) enrolled patients with confirmed N2 EGFR mutated NSCLC. They received erlotinib for 6 weeks preoperatively followed by 1 year postoperatively, or chemotherapy (cisplatin-gemcitabine) for two cycles preoperatively and two cycles postoperatively that. Radiological response rates and RFS favored erlotinib, but there were no pathological complete responses and major pathological responses (less than 10% tumor remaining at resection) were 10% in the erlotinib group. There were no differences in OS (67,68).
Two single-arm clinical trials have been reported with osimertinib. The NEOS trial (ChiCTR-1800016948) evaluated 6 weeks of treatment before surgery in 88 patients; radiological responses were encouraging but pathological responses were only 15% (4% major and 11% complete responses) (69). The UCSF trial evaluated treatment with osimertinib for 4–8 weeks in 27 patients and reported a major pathological response rate of 15% (70).
The phase 3 randomized NeoADAURA clinical trial is also ongoing (NCT-04351555), comparing osimertinib versus osimertinib with chemotherapy versus chemotherapy for 9 weeks prior to surgery. There are other ongoing phase 2 trials with first generation TKIs (icotinib monotherapy, NCT-03749213, or with chemotherapy, NCT-05104788, NeoIpower trial), with second generation TKIs (afatinib, NCT-04201756) and with third generation TKIs (aumolertinib, NCT-04455594, Answer trial).
Conclusions
EGFR mutant early-stage NSCLC has a variable incidence according to geographical location, and its clinical and molecular features are diverse. Determination of EGFR mutation status in early-stage patients is necessary for optimal therapeutic decision-making, as the presence of an EGFR mutation implies lack of benefit from adjuvant or neoadjuvant immunotherapy and EGFR-TKIs are available. In fact, adjuvant treatment with osimertinib for 3 years after surgery, with or without chemotherapy, has improved RFS and OS in these patients, without compromising quality of life.
Acknowledgments
Funding: None.
Footnote
Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://cco.amegroups.com/article/view/10.21037/cco-24-35/rc
Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-24-35/prf
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-24-35/coif). S.M.R. reports taking part as an invited speaker for Sanofi, BMS and Takeda; receiving support from Merck, Sanofi, Lilly, BMS, and Pfizer for meeting registration, accommodation and/or travel expenses, all outside the present manuscript. A.B. reports taking part as invited speaker for BMS, MSD, Pfizer, Sanofi, Pierre Fabre, Novartis, Takeda and Astra Zeneca; receiving support from BMS, MSD, Sanofi, Pfizer and Astra Zeneca for meeting registration, accommodation and/or travel expenses; and taking part in an Advisory Board for BMS, MSD and Sanofi, all outside the present manuscript. M.M. reports receiving grants from Roche and Astra Zeneca; taking part as invited speaker for Roche, MSD, Pfizer, Sanofi, Pierre Fabre, Novartis, Takeda and Astra Zeneca; and receiving support for attending meetings and/or travel from Astra Zeneca, MSD, Sanofi, all outside the present manuscript. The authors have no other conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Sharma SV, Bell DW, Settleman J, et al. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 2007;7:169-81. [Crossref] [PubMed]
- D'Angelo SP, Pietanza MC, Johnson ML, et al. Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol 2011;29:2066-70. [Crossref] [PubMed]
- Kawaguchi T, Matsumura A, Fukai S, et al. Japanese ethnicity compared with Caucasian ethnicity and never-smoking status are independent favorable prognostic factors for overall survival in non-small cell lung cancer: a collaborative epidemiologic study of the National Hospital Organization Study Group for Lung Cancer (NHSGLC) in Japan and a Southern California Regional Cancer Registry databases. J Thorac Oncol 2010;5:1001-10. [Crossref] [PubMed]
- Shi Y, Au JS, Thongprasert S, et al. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER). J Thorac Oncol 2014;9:154-62. [Crossref] [PubMed]
- Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947-57. [Crossref] [PubMed]
- Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011;12:735-42. [Crossref] [PubMed]
- Park K, Tan EH, O'Byrne K, et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol 2016;17:577-89. [Crossref] [PubMed]
- Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:113-25. [Crossref] [PubMed]
- Mok TS, Cheng Y, Zhou X, et al. Updated Overall Survival in a Randomized Study Comparing Dacomitinib with Gefitinib as First-Line Treatment in Patients with Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. Drugs 2021;81:257-66. [Crossref] [PubMed]
- Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N Engl J Med 2020;382:41-50. [Crossref] [PubMed]
- Beau-Faller M, Prim N, Ruppert AM, et al. Rare EGFR exon 18 and exon 20 mutations in non-small-cell lung cancer on 10 117 patients: a multicentre observational study by the French ERMETIC-IFCT network. Ann Oncol 2014;25:126-31. [Crossref] [PubMed]
- Penzel R, Sers C, Chen Y, et al. EGFR mutation detection in NSCLC--assessment of diagnostic application and recommendations of the German Panel for Mutation Testing in NSCLC. Virchows Arch 2011;458:95-8. [Crossref] [PubMed]
- Surveillance Research Program, National Cancer Institute. SEER*Explorer: An interactive website for SEER cancer statistics [Internet] [Internet]. [cited 2024 Jan 2]. Available online: https://seer.cancer.gov/statistics-network/explorer/application.html
- Mordant P MD. Outcome of Patients With Resected Early-Stage Non-small Cell Lung Cancer and EGFR Mutations: Results From the IFCT Biomarkers France Study. Clin Lung Cancer 2023;24:1-10. [Crossref] [PubMed]
- Hondelink LM, Ernst SM, Atmodimedjo P, et al. Prevalence, clinical and molecular characteristics of early stage EGFR-mutated lung cancer in a real-life West-European cohort: Implications for adjuvant therapy. Eur J Cancer 2023;181:53-61. [Crossref] [PubMed]
- Pi C, Xu CR, Zhang MF, et al. EGFR mutations in early-stage and advanced-stage lung adenocarcinoma: Analysis based on large-scale data from China. Thorac Cancer 2018;9:814-9. [Crossref] [PubMed]
- Suda K, Mitsudomi T, Shintani Y, et al. Clinical Impacts of EGFR Mutation Status: Analysis of 5780 Surgically Resected Lung Cancer Cases. Ann Thorac Surg 2021;111:269-76. [Crossref] [PubMed]
- Saw SPL, Zhou S, Chen J, et al. Association of Clinicopathologic and Molecular Tumor Features With Recurrence in Resected Early-Stage Epidermal Growth Factor Receptor-Positive Non-Small Cell Lung Cancer. JAMA Netw Open 2021;4:e2131892. [Crossref] [PubMed]
- Kosaka T, Yatabe Y, Onozato R, et al. Prognostic implication of EGFR, KRAS, and TP53 gene mutations in a large cohort of Japanese patients with surgically treated lung adenocarcinoma. J Thorac Oncol 2009;4:22-9. [Crossref] [PubMed]
- Takamochi K, Oh S, Matsunaga T, et al. Prognostic impacts of EGFR mutation status and subtype in patients with surgically resected lung adenocarcinoma. J Thorac Cardiovasc Surg 2017;154:1768-1774.e1. [Crossref] [PubMed]
- Wu YL, Tsuboi M, He J, et al. Osimertinib in Resected EGFR-Mutated Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:1711-23. [Crossref] [PubMed]
- Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 2021;398:1344-57. [Crossref] [PubMed]
- Ginsberg RJ, Rubinstein LV. Randomized trial of lobectomy versus limited resection for T1 N0 non-small cell lung cancer. Lung Cancer Study Group. Ann Thorac Surg 1995;60:615-22; discussion 622-3. [Crossref] [PubMed]
- Rosen JE, Keshava HB, Yao X, et al. The Natural History of Operable Non-Small Cell Lung Cancer in the National Cancer Database. Ann Thorac Surg 2016;101:1850-5. [Crossref] [PubMed]
- Saji H, Okada M, Tsuboi M, et al. Segmentectomy versus lobectomy in small-sized peripheral non-small-cell lung cancer (JCOG0802/WJOG4607L): a multicentre, open-label, phase 3, randomised, controlled, non-inferiority trial. Lancet 2022;399:1607-17. [Crossref] [PubMed]
- Altorki N, Wang X, Kozono D, et al. Lobar or Sublobar Resection for Peripheral Stage IA Non-Small-Cell Lung Cancer. N Engl J Med 2023;388:489-98. [Crossref] [PubMed]
- Pignon JP, Tribodet H, Scagliotti GV, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol 2008;26:3552-9. [Crossref] [PubMed]
- Postmus PE, Kerr KM, Oudkerk M, et al. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2017;28:iv1-iv21. [Crossref] [PubMed]
- Kris MG, Gaspar LE, Chaft JE, et al. Adjuvant Systemic Therapy and Adjuvant Radiation Therapy for Stage I to IIIA Completely Resected Non-Small-Cell Lung Cancers: American Society of Clinical Oncology/Cancer Care Ontario Clinical Practice Guideline Update. J Clin Oncol 2017;35:2960-74. [Crossref] [PubMed]
- NCCN Clinical Practice Guideliness in Oncology. NSCLC guidelines, version 1.2024 [Internet]. [cited 2024 Jan 3]. Available online: https://www.nccn.org/guidelines/guidelines-detail
- Lim E, Harris G, Patel A, et al. Preoperative versus postoperative chemotherapy in patients with resectable non-small cell lung cancer: systematic review and indirect comparison meta-analysis of randomized trials. J Thorac Oncol 2009;4:1380-8. [Crossref] [PubMed]
- Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet 2014;383:1561-71. [Crossref] [PubMed]
- Lee CK, Man J, Lord S, et al. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non-Small Cell Lung Cancer-A Meta-Analysis. J Thorac Oncol 2017;12:403-7. [Crossref] [PubMed]
- Garassino MC, Gelibter AJ, Grossi F, et al. Italian Nivolumab Expanded Access Program in Nonsquamous Non-Small Cell Lung Cancer Patients: Results in Never-Smokers and EGFR-Mutant Patients. J Thorac Oncol 2018;13:1146-55. [Crossref] [PubMed]
- Mazieres J, Drilon A, Lusque A, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 2019;30:1321-8. [Crossref] [PubMed]
- Yang JCH, Lee DH, Lee JS, et al. Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: Phase 3 KEYNOTE-789 study. J Clin Oncol 2023;41:LBA9000. [Crossref]
- Mok T, Nakagawa K, Park K, et al. Nivolumab Plus Chemotherapy in Epidermal Growth Factor Receptor-Mutated Metastatic Non-Small-Cell Lung Cancer After Disease Progression on Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors: Final Results of CheckMate 722. J Clin Oncol 2024;42:1252-64. [Crossref] [PubMed]
- O'Brien M, Paz-Ares L, Marreaud S, et al. Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer (PEARLS/KEYNOTE-091): an interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol 2022;23:1274-86. [Crossref] [PubMed]
- Provencio M, Nadal E, Insa A, et al. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol 2020;21:1413-22. [Crossref] [PubMed]
- Provencio M, Nadal E, González-Larriba JL, et al. Perioperative Nivolumab and Chemotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 2023;389:504-13. [Crossref] [PubMed]
- Forde PM, Spicer J, Lu S, et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N Engl J Med 2022;386:1973-85. [Crossref] [PubMed]
- Cascone T, Awad MM, Spicer JD, et al. LBA1 CheckMate 77T: Phase III study comparing neoadjuvant nivolumab (NIVO) plus chemotherapy (chemo) vs neoadjuvant placebo plus chemo followed by surgery and adjuvant NIVO or placebo for previously untreated, resectable stage II–IIIb NSCLC. Ann Oncol 2023;34:S1295. [Crossref]
- Heymach JV, Harpole D, Mitsudomi T, et al. Perioperative Durvalumab for Resectable Non-Small-Cell Lung Cancer. N Engl J Med 2023;389:1672-84. [Crossref] [PubMed]
- Wakelee H, Liberman M, Kato T, et al. Perioperative Pembrolizumab for Early-Stage Non-Small-Cell Lung Cancer. N Engl J Med 2023;389:491-503. [Crossref] [PubMed]
- Remon J, Soria JC, Peters S, et al. Early and locally advanced non-small-cell lung cancer: an update of the ESMO Clinical Practice Guidelines focusing on diagnosis, staging, systemic and local therapy. Ann Oncol 2021;32:1637-42. [Crossref] [PubMed]
- Pisters K, Kris MG, Gaspar LE, et al. Adjuvant Systemic Therapy and Adjuvant Radiation Therapy for Stage I-IIIA Completely Resected Non-Small-Cell Lung Cancer: ASCO Guideline Rapid Recommendation Update. J Clin Oncol 2022;40:1127-9. [Crossref] [PubMed]
- Kelly K, Altorki NK, Eberhardt WE, et al. Adjuvant Erlotinib Versus Placebo in Patients With Stage IB-IIIA Non-Small-Cell Lung Cancer (RADIANT): A Randomized, Double-Blind, Phase III Trial. J Clin Oncol 2015;33:4007-14. [Crossref] [PubMed]
- Zhong WZ, Wang Q, Mao WM, et al. Gefitinib Versus Vinorelbine Plus Cisplatin as Adjuvant Treatment for Stage II-IIIA (N1-N2) EGFR-Mutant NSCLC: Final Overall Survival Analysis of CTONG1104 Phase III Trial. J Clin Oncol 2021;39:713-22. [Crossref] [PubMed]
- Tada H, Mitsudomi T, Misumi T, et al. Randomized Phase III Study of Gefitinib Versus Cisplatin Plus Vinorelbine for Patients With Resected Stage II-IIIA Non-Small-Cell Lung Cancer With EGFR Mutation (IMPACT). J Clin Oncol 2022;40:231-41. [Crossref] [PubMed]
- Yue D, Xu SD, Wang Q, et al. Updated overall survival (OS) and exploratory analysis from the randomized, phase II EVAN study of erlotinib (E) versus vinorelbine plus cisplatin (NP) as adjuvant therapy in Chinese patients with stage IIIA EGFR + NSCLC. J Clin Oncol 2021;39:8520. [Crossref]
- He J, Su C, Liang W, et al. Icotinib versus chemotherapy as adjuvant treatment for stage II-IIIA EGFR-mutant non-small-cell lung cancer (EVIDENCE): a randomised, open-label, phase 3 trial. Lancet Respir Med 2021;9:1021-9. [Crossref] [PubMed]
- Ou W, Li N, Wang BX, et al. Adjuvant icotinib versus observation in patients with completely resected EGFR-mutated stage IB NSCLC (GASTO1003, CORIN): a randomised, open-label, phase 2 trial. EClinicalMedicine 2023;57:101839. [Crossref] [PubMed]
- Cheng H, Li XJ, Wang XJ, et al. A meta-analysis of adjuvant EGFR-TKIs for patients with resected non-small cell lung cancer. Lung Cancer 2019;137:7-13. [Crossref] [PubMed]
- Tang W, Li X, Xie X, et al. EGFR inhibitors as adjuvant therapy for resected non-small cell lung cancer harboring EGFR mutations. Lung Cancer 2019;136:6-14. [Crossref] [PubMed]
- Lv C, Wang R, Li S, et al. Randomized phase II adjuvant trial to compare two treatment durations of icotinib (2 years versus 1 year) for stage II-IIIA EGFR-positive lung adenocarcinoma patients (ICOMPARE study). ESMO Open 2023;8:101565. [Crossref] [PubMed]
- Herbst RS, Wu YL, John T, 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. J Clin Oncol 2023;41:1830-40. [Crossref] [PubMed]
- Majem M, Goldman JW, John T, et al. Health-Related Quality of Life Outcomes in Patients with Resected Epidermal Growth Factor Receptor-Mutated Non-Small Cell Lung Cancer Who Received Adjuvant Osimertinib in the Phase III ADAURA Trial. Clin Cancer Res 2022;28:2286-96. [Crossref] [PubMed]
- Wu YL, John T, Grohe C, et al. Postoperative Chemotherapy Use and Outcomes From ADAURA: Osimertinib as Adjuvant Therapy for Resected EGFR-Mutated NSCLC. J Thorac Oncol 2022;17:423-33. [Crossref] [PubMed]
- Tsuboi M, Herbst RS, John T, et al. Overall Survival with Osimertinib in Resected EGFR-Mutated NSCLC. N Engl J Med 2023;389:137-47. [Crossref] [PubMed]
- Reungwetwattana T, Nakagawa K, Cho BC, et al. CNS Response to Osimertinib Versus Standard Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients With Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2018; Epub ahead of print. [Crossref] [PubMed]
- Saito R, Sugawara S, Ko R, et al. Phase 2 study of osimertinib in combination with platinum and pemetrexed in patients with previously untreated EGFR-mutated advanced non-squamous non-small cell lung cancer: The OPAL Study. Eur J Cancer 2023;185:83-93. [Crossref] [PubMed]
- Planchard D, Jänne PA, Cheng Y, et al. Osimertinib with or without Chemotherapy in EGFR-Mutated Advanced NSCLC. N Engl J Med 2023;389:1935-48. [Crossref] [PubMed]
- Ahn MJ, Jung HA, Ku BM, et al. 933MO Longitudinal monitoring of circulating tumor DNA from plasma in patients with curative resected stage IA-IIIA EGFR mutant non-small cell lung cancer. Ann Oncol 2022;33:S427-S437. [Crossref]
- Wang Z, Cheng Y, An T, et al. Detection of EGFR mutations in plasma circulating tumour DNA as a selection criterion for first-line gefitinib treatment in patients with advanced lung adenocarcinoma (BENEFIT): a phase 2, single-arm, multicentre clinical trial. Lancet Respir Med 2018;6:681-90. [Crossref] [PubMed]
- Liu SY, Bao H, Wang Q, et al. Genomic signatures define three subtypes of EGFR-mutant stage II-III non-small-cell lung cancer with distinct adjuvant therapy outcomes. Nat Commun 2021;12:6450. [Crossref] [PubMed]
- Saw SPL, Ng WP, Zhou S, et al. PD-L1 score as a prognostic biomarker in asian early-stage epidermal growth factor receptor-mutated lung cancer. Eur J Cancer 2023;178:139-49. [Crossref] [PubMed]
- Zhong WZ, Chen KN, Chen C, et al. Erlotinib Versus Gemcitabine Plus Cisplatin as Neoadjuvant Treatment of Stage IIIA-N2 EGFR-Mutant Non-Small-Cell Lung Cancer (EMERGING-CTONG 1103): A Randomized Phase II Study. J Clin Oncol 2019;37:2235-45. [Crossref] [PubMed]
- Wu YL, Zhong W, Chen KN, et al. CTONG1103: Final overall survival analysis of the randomized phase 2 trial of erlotinib versus gemcitabine plus cisplatin as neoadjuvant treatment of stage IIIA-N2 EGFR-mutant non–small cell lung cancer. J Clin Oncol 2021;39:8502. [Crossref]
- Lv C, Fang W, Wu N, et al. Osimertinib as neoadjuvant therapy in patients with EGFR-mutant resectable stage II-IIIB lung adenocarcinoma (NEOS): A multicenter, single-arm, open-label phase 2b trial. Lung Cancer 2023;178:151-6. [Crossref] [PubMed]
- Blakely C, Urisman A, Kerr D, et al. P26.02. A Phase II Trial of Neoadjuvant Osimertinib for Surgically Resectable EGFR-Mutant Non-Small Cell Lung Cancer: Updated Results. J Thorac Oncol 2021;16:S1039-40. [Crossref]