| Home | E-Submission | Sitemap | Contact Us |  
top_img
Cancer Research and Treatment > Volume 55(4); 2023 > Article
Kim, Jang, Choi, Kim, Lee, Chang, Lee, Kim, Yang, Ryu, Lee, Lee, Park, Lee, Jang, Yoon, and Oh: Final Report on Real-World Effectiveness of Sequential Afatinib and Osimertinib in EGFR-Positive Advanced Non–Small Cell Lung Cancer: Updated Analysis of the RESET Study

Abstract

Purpose

This study aimed to report the final analysis of time-on-treatment (TOT) and overall survival (OS) in patients with advanced-stage epidermal growth factor receptor (EGFR)+ non–small cell lung cancer (NSCLC) who received sequential afatinib and osimertinib and to compare the outcomes with other second-line regimens (comparator group).

Materials and Methods

In this updated report, the existing medical records were reviewed and rechecked. TOT and OS were updated and analyzed according to clinical features using the Kaplan-Meier method and log-rank test. TOT and OS were compared with those of the comparator group, in which most patients received pemetrexed-based treatments. A multivariable Cox proportional hazard model was used to evaluate features that could affect survival outcomes.

Results

The median observation time was 31.0 months. The follow-up period was extended to 20 months. A total of 401 patients who received first-line afatinib were analyzed (166 with T790M+ and second-line osimertinib, and 235 with unproven T790M and other second-line agents). Median TOTs on afatinib and osimertinib were 15.0 months (95% confidence interval [CI], 14.0 to 16.1) and 11.9 months (95% CI, 8.9 to 14.6), respectively. The median OS in the osimertinib group was 54.3 months (95% CI, 46.7 to 61.9), much longer than that in the comparator group. In patients who received osimertinib, the OS was longest with Del19+ (median, 59.1; 95% CI, 48.7 to 69.5).

Conclusion

This is one of the largest real-world studies reporting the encouraging activity of sequential afatinib and osimertinib in Asian patients with EGFR+ NSCLC who acquired the T790M mutation, particularly Del19+.

Introduction

Lung cancer is the leading cause of death worldwide, accounting for almost 22% of all cancer–related deaths in males and 14% in females [1]. There is an increasing trend in lung cancer incidence in Eastern Asia, especially in the female population [2]. Although the most common histological type of lung cancer may vary between countries, adenocarcinoma is currently more prevalent than squamous cell carcinoma [3]. The incidence of adenocarcinoma in females continues to rise in several countries, while it remains stable in males [4]. A recent study in South Korea reported an increasing rate of adenocarcinoma and a decreasing trend of squamous cell carcinoma [5].
The selection of treatment strategy is an integral part of cancer management because it can lead to a significant improvement in survival outcomes. Tyrosine kinase inhibitors (TKIs), such as erlotinib, gefitinib, and afatinib, have been the mainstay for the management of advanced non–small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations. The FLAURA phase 3 trial recommended single-agent osimertinib as the preferred first-line treatment for advanced EGFR+ NSCLC [6]. However, the question remains whether osimertinib should be administered as the first- or second-line treatment following first- or second-generation TKIs.
The T790M mutation is the most common resistance mechanism to first-generation (erlotinib and gefitinib) and second-generation (afatinib) TKIs during first-line treatment [7]. In the AURA3 trial, osimertinib as a second-line treatment demonstrated a striking effect against the T790M mutation [8]. Given that post-osimertinib treatment is challenging and the drug is not reimbursable as a first-line treatment in some countries such as South Korea, many clinicians would reserve osimertinib for T790M positive progression.
At present, few studies, including randomized controlled trials and real-world reports, are available on the activity of sequential TKI treatments in EGFR+ advanced NSCLC. For example, a post-hoc analysis of the LUX-Lung 7 study reported a 3-year survival rate of greater than 90% in patients who received sequential afatinib and osimertinib [9]. GioTag and UpSwinG are other studies showing the effectiveness of sequential afatinib and osimertinib therapies in real-world practice [10,11]. However, these studies might be limited by the small number of Asian patients and lack of a comparator group. Given the ethnic differences in the clinical effects of EGFR-TKIs, there is still a paucity of data on sequential afatinib and osimertinib treatment in Asian populations.
The real-world multicenter RESET study from South Korea might have provided an insight into the optimal sequence of EGFR-TKIs in Asian populations by investigating more than 700 advanced EGFR+ NSCLC patients [12]. The preliminary results were promising with time-on-treatment (TOT) on first-line afatinib of 15.7 months and TOT on second-line osimertinib treatment for 11.9 months. Overall survival (OS) was not achieved in patients receiving afatinib and subsequent osimertinib treatment. Many patients were still on the afatinib and osimertinib treatment. Therefore, it would be worthwhile to report updated patient data in the RESET study.

Materials and Methods

1. Datasets and patient selection

The design of the RESET study has been described previously [12]. In brief, RESET was a retrospective observational study conducted across 16 medical centers in South Korea. This study was designed to evaluate the real-world effectiveness of sequential afatinib and osimertinib treatments in patients with advanced EGFR+ NSCLC. Currently, osimertinib is the only approved EGFR-TKI after the failure of first-line TKIs in patients in South Korea. In a previous report, 164 patients were still receiving treatment, and 56 patients continued osimertinib at the final follow-up. Therefore, we expanded the final follow-up date from October 2020 to 30 June 2022 and collected the updated survival outcome for those patients. In addition, we rechecked the information on the clinical, molecular, and histologic features as well as the data regarding treatment outcomes, such as the date of treatment initiation and discontinuation and the occurrence of treatment-related events.
The data-processing flow is illustrated in Fig. 1. The original cohort dataset comprised 735 patients. In the first selection process, 289 patients were excluded for the following reasons: 54 continued afatinib, 12 had unavailable updated data, 77 were transferred or lost to follow-up, and 148 had no data for second-line treatment. In the next step, in which patients had information on both first- and second-line treatment, 45 patients were excluded: 26 received osimertinib without evidence of T790M and 19 were administered drugs other than osimertinib despite having T790M+. Consistent with a previous report, we classified the patients according to whether they received osimertinib as a second-line treatment. All patients in the osimertinib group 100% presented with the T790M mutation after afatinib failure. All patients in the comparator group were negative or unproven for T790M and received other therapies.

2. Variables

In our previous report, we investigated features, including age, sex, Eastern Cooperative Oncology Group (ECOG) performance status (PS), smoking status (never, former, and current), tissue type of NSCLC at the initial diagnosis (adenocarcinoma or others), type and presence of EGFR mutation (deletion 19 [Del19], L858R, and others), which was detected using the peptic nucleic acid-mediated real-time polymerase chain reaction clamping method (Panagene, Daejeon, Korea) or the Roche Cobas EGFR mutation test (Roche Molecular Systems, Pleasanton, CA), tumor stage assessed by the eighth edition of the American Joint Committee on Cancer (AJCC) stating manual, number of metastatic organs, presence of metastasis in specific organs, type of brain metastasis at the initial work-up, change in brain metastasis during afatinib, and dose adjustment for afatinib. In addition, we also collected information on anthropometric indices, such as height and weight, longest tumor diameter, and TNM stage at the initial diagnosis.

3. Outcomes

The analysis of treatment-related outcomes was exploratory. The primary purpose of this updated report was to expand the final follow-up period to 20 months, as mentioned above. OS was defined as the length of time from the initiation of first-line afatinib therapy to death from any cause. TOT was also updated; the period was estimated for first-line afatinib and second-line osimertinib or other therapies, separately. TOT was defined as the period between the start of treatment and discontinuation of the drug for any reason, including tumor progression, drug toxicity, or death.

4. Statistics

Clinical characteristics were summarized as numbers with percentages for categorical variables and means with standard deviations for continuous variables. Clinical features were compared using chi-square test or Fisher’s exact test for categorical features and Student’s t test for continuous variables.
TOT and OS were estimated and visualized using the Kaplan-Meier method. The log-rank test was used to compare the differences between survival outcomes within the categorical variables. The median period (months) and 95% confidence interval (CI) were also measured. TOT and OS were updated in both the afatinib→osimertinib and comparator groups.
Additionally, a subgroup analysis was performed by stratifying the types of second-line treatments other than osimertinib. At the time of primary data collection, we observed that most of the patients received pemetrexed alone or in combination with platinum-based agents as second-line agents. OS was compared between osimertinib vs. pemetrexed-containing regimens, and osimertinib vs. pemetrexed-platinum doublet vs. pemetrexed monotherapy.
The Cox proportional hazard model was used to identify the features that could affect TOT and OS. Multivariable analyses were performed using factors with p < 0.1 in the univariable model.
All statistical analyses were performed using R software ver. 4.2.2 for Windows (R Development Core Team, R Foundation for Statistical Computing, Vienna, Austria) and IBM SPSS Statistics for Windows ver. 25.0 (IBM Corp., Armonk, NY).

Results

A total of 401 patients across 16 medical centers were included in the analysis. The median observation time was 31.0 months (interquartile range, 19.8 to 45.9). The male and female patients were equally distributed (Table 1). Patients who received osimertinib as second-line treatment had a higher AJCC stage than those who received other therapies. The rate of Del19+ cells was higher in the osimertinib-treated group. Patients who received osimertinib had a higher rate of recurrence or new detection of brain metastasis more frequently than those in the comparator group.
The median TOT during afatinib was 15.0 months (95% CI, 14.0 to 16.1) (Table 2, S1 Fig.). Median TOT during afatinib was 16.6 (95% CI, 15.2 to 18.0) in the osimertinib-treated group and 13.9 (95% CI, 12.4 to 15.3) in the comparator group with p of 0.043 (data not shown). The TOT during osimertinib was 11.9 months (95% CI, 8.9 to 14.6) (Table 2, S2 Fig.), which was significantly longer than that in patients who received other treatments (5.1 months; 95% CI, 4.2 to 5.9) with a p-value of < 0.001. TOT during afatinib treatment was longer in patients with a Del19 mutation (15.7 months; 95% CI, 14.1 to 17.3) than in patient with a L858R mutation or other mutations (p=0.037). However, the period did not differ significantly between the types of mutations during osimertinib treatment.
Updated median OS in all 401 patients were estimated as 49.1 months (95% CI, 43.6 to 54.6). The median OS in patients who received sequential afatinib and osimertinib was 54.3 months (95% CI, 46.7 to 61.9) (Table 3, Fig. 2). The OS was longer in patients who received osimertinib as second-line treatment than in patients who received other regiments (41.3 months; 95% CI, 32.9 to 49.8; p=0.019). OS was the longest in patients with a Del19 mutation (59.1 months; 95% CI, 48.7 to 69.5).
The multivariable Cox proportional hazard model showed that poor ECOG PS, histologic types other than adenocarcinoma, EGFR mutations other than Del19 and L858R, higher numbers of metastatic organs, and no dose adjustment during afatinib treatment were related to an increased risk of poor TOT (Table 4). Meanwhile, in terms of OS, the hazard ratio was higher in patients with poor PS and the presence of liver metastasis during afatinib and osimertinib treatments (Table 5). In patients receiving afatinib followed by other regimens, other types of EGFR mutations, liver metastasis, and no dose adjustment during first-line afatinib were associated with a decrease in OS.
The types of second-line agents other than osimertinib in the comparator groups are summarized in S3 Table. As noted above, most patients (n=146, 66.7%) were treated with pemetrexed-based treatments: 74 (33.3%) received pemetrexed-platinum doublet and 74 (33.3%) received pemetrexed monotherapy. Patients who received sequential afatinib and osimertinib showed longer OS (median, 54.3 months; 95% CI, 48.5 to not available [NA]) than those who received pemetrexed-containing regimens (median, 41.7 months; 95% CI, 36.7 to 67.3; p=0.039) for 12.6 months numerically (Fig. 3, S4 Table). In the comparator group, pemetrexed-platinum doublet therapy showed longer OS than pemetrexed monotherapy, although both regimens conferred shorter OS than the osimertinib group (p=0.005) (Fig. 4, S4 Table). However, when comparing patients administered osimertinib with pemetrexed-platinum doublet therapy, a statistical significance was not reached (p=0.6), although median OS was numerically longer in osimertinib group of 54.3 months than in doublet group (50.0 months; 95% CI, 37.6 to NA).

Discussion

The RESET study is the first multicenter study in South Korea to report survival outcomes in patients with advanced EGFR+ NSCLC who received sequential afatinib and osimertinib treatment. All patients who received osimertinib as a second-line treatment were all T790M positive. This study has several strengths. First, this final analysis of RESET is one of the largest studies in Asian populations analyzing the real-world effectiveness of osimertinib after afatinib treatment. Second, the RESET study brought the comparator group into the survival outcome analysis, which was absent in other real-world studies. Sequential afatinib and osimertinib were superior to other agents, mostly pemetrexed-based treatments, although the presence of the T790M mutation is the key to deciding the second-line treatment. Third, the survival outcomes were subject to comprehensive analyses based on various clinical factors. The encouraging activity of sequential afatinib and osimertinib in real-world data supports the feasibility of applying this treatment sequence in clinical practice by reserving osimertinib as a second-line regimen. Further scrutiny of prospective clinical trials is required to apply our results to real-world clinical practice. For example, a randomized open-label phase 4 trial in Germany, AFAMOSI, is going to evaluate the efficacy of afatinib followed by osimertinib in treatment-naïve patients with EGFR+ and T790M non-squamous NSCLC (NCT04413201).
The primary objective of RESET is to report the updated OS. The median OS was not reached in our previous study, and the updated median OS herein was 54.3 months. In terms of sequential afatinib and osimertinib treatment, only a few prospective studies have reported OS in patients with EGFR + NSCLC who received TKIs in that sequence. For example, median OS was ‘not evaluable’ with afatinib versus 46.0 months with gefitinib in patients who received the following osimertinib in a sub-analysis of the LUX-Lung 7 trial [9]. However, only 20 patients who were treated with afatinib received osimertinib as second-line treatment, and 23 patients treated with gefitinib received osimertinib.
In this final report, the estimated median TOT was 15.0 months on afatinib and 11.9 months on osimertinib. The results from previous randomized controlled trials substantiate the findings of RESET. The median duration of afatinib was 13.7 months in the post-hoc analysis of the LUX-Lung 7 trial [9]. First-line afatinib in the Asian population demonstrated a median progression-free survival of 11.0 months in the LUX-Lung 6 trial [13]. For osimertinib, the results from the phase 3 AURA trial showed that the median period of progression-free survival was 10.1 months in patients who received the drug after disease progression with first-line TKIs [8]. However, only 20 patients (7%) were treated with afatinib before osimertinib treatment. A subgroup analysis of the AURA 3 study in 63 Japanese patients showed slightly longer period of progression-free survival of 12.5 months [14].
Interestingly, patients who received pemetrexed-platinum doublet therapy as a second-line treatment had a longer OS period than those who received pemetrexed monotherapy. This observation does not necessarily imply that combination therapy is superior to monotherapy. Patients in the former were 3.8 years younger than those in the latter, and the proportion of patients with ECOG PS ≥ 2 was 12.3% in the combination group versus 6.7% in the monotherapy group. A phase 2 randomized clinical trial comparing the efficacy of pemetrexed-carboplatin doublet versus pemetrexed monotherapy as a second-line treatment in patients with advanced NSCLC yielded similar findings [15]. Patients who received combination therapy had a significantly longer progression-free survival.
Given the lack of data from prospective trials, evidence from real-world practice may provide additional insights into the optimization of treatment sequences within TKIs. Currently, except for RESET, two global multinational observational studies are available. The UpSwinG study enrolled 191 patients across nine countries with advanced EGFR+ NSCLC who were treated with first-line afatinib, following the detection of T790M, and second-line osimertinib [11]. The study analyzed 118 Asians, whereas our study included 166 South Koreans. In the UpSwinG study, the median OS in Asian patients was 42.3 months (95% CI, 33.2 to 63.5), which is slightly shorter than the OS in our study. Another global, multinational non-interventional study was GioTag [10]. However, GioTag only involved 50 Asian patients with a median OS of 44.8 months (95% CI, 37.0 to 57.8).
These real-world experiences show that the survival benefit is particularly promising in Asian patients with Del19+ disease. In GioTag study, the median OS in Asians with Del19+ versus all patients with Del19+ was 44.8 months (90% CI, 37.0 to 57.8) versus 41.6 months (90% CI, 36.9 to 45.0) [10]. A combined analysis of GioTag and UpSwinG studies showed that the median OS in Asian patients were significantly different between Del19 (n=109) and L858R (n=59) mutations; 63.5 months (95% CI, 42.3 to 71.1) and 39.1 (95% CI, 29.3 to 48.5), respectively [16]. These findings are comparable with the results from the updated RESET report, where 98 patients tested positive for Del19 and 55 were positive for L858R (Table 6). In a group received sequential afatinib and osimertinib therapy, the median OS was 59.1 months for the Del19+ and 46.5 months for L858R+ mutation. Consequently, our data support the notion that sequential afatinib treatment followed by osimertinib is an effective therapeutic option in Asian patients with advanced EGFR+ NSCLC, especially those with Del19+.
In addition, the authors of the above two real-world studies noted that they were largely limited by the lack of comparator arms. In this regard, our study additionally investigated specific regimens that were used as second-line treatments. Numerically, patients treated with osimertinib showed a 13.0-month extension of OS compared to the comparator group and 12.6-month extension to the pemetrexed-containing treatments. However, it should be noted that the T790M mutation was not detected or unproved in the comparator group. Several studies have reported similar results for the RESET. Progression-free survival was longer in patients with T790M+ NSCLC after initial TKI failure than in patients with T790M negativity [17]. In addition to second-line treatment, T790M mutation expression is associated with indolent progression during first-line treatment with both TKIs and chemotherapy agents [18]. These clinical results were further supported by experimental models. Cells harboring the T790M mutation showed a slower rate of growth in a preclinical study [19]. Mice expressing T790M showed a longer latency to tumorigenesis than those expressing other EGFR mutations [20].
Currently, there are no approved targeted treatments for patients who experience disease progression after osimertinib treatment. Platinum-based doublet chemotherapy may be the next step for these patients. A phase 1/1b Chrysalis-2 trial is an effort to examine post-osimertinib treatments using lazertinib as monotherapy or in combination with amivantamab (NCT04077463). A substantial proportion of patients receiving osimertinib develop resistance, despite a durable response. Especially, C797S point mutation in exon 20 is particularly important for osimertinib resistance [21], accounting for 10%–26% of cases of resistance after second-line osimertinib [8]. However, TKI resistance mechanisms may differ in the presence of osimertinib or afatinib. In an in vitro examination, mutations developed differently between cancer cells exposed to either osimertinib or afatinib [22]. Another preclinical examination support that a combination of osimertinib and afatinib rather than either drug alone was more effective in an appearance of drug-resistant cells [22].
The controversy regarding the optimal sequence of osimertinib may be intensified by the results of the FLAURA study: an updated OS in patients received first-line osimertinib was 38.6 months in the first-line osimertinib group [23], which was much shorter than the RESET. There could be several explanations around this difference. First, while 68% of patients who received osimertinib as a first-line treatment in the FLAURA study administered cytotoxic chemotherapy as a second-line treatment [23], patients in RESET study received two subsequent EGFR-TKIs, afatinib followed by osimertinib. Second, the medical environment and a health-care system in South Korea is generally considered to be advanced and of a high standard. According to a study of cancer statistics published in South Korea, cancer survival rates were generally higher than those in other countries [24]. Third, as we have shown in Table 6, survival data of RESET study was comparable to the previous real-world studies. Although our results are based on a retrospective design, relatively enough patients were analyzed, and survival periods such as TOT and OS were comprehensively estimated according to various clinical features. Our findings suggest that sequential therapy with afatinib followed by osimertinib is effective and could potentially become an option for patients with advanced EGFR+ NSCLC.
Despite the strengths of our study, because of the inherent nature of the retrospective study, it has several limitations, as noted in a previous report [12]. Selection bias could exist, since this study was restricted to South Korea, where osimertinib is reimbursable only for patients in whom first-line EGFR-TKI failed and T790M upon re-biopsy subsequently tested positive, and this issue could not be corrected. Further studies investigating the survival outcomes after first- and second-line osimertinib treatment would be valuable, given that osimertinib is the preferred first-line treatment option for advanced EGFR+ NSCLC based on the FLAURA study findings [7]. Misclassification may also occur. We attempted to minimize this problem by reviewing and rechecking the collected data. Survival data were not mature in our previous report, but we expanded the observation period up to 20 months. Another study limitation was the lack of data on adverse events, which might have affected the accuracy and completeness of our findings regarding drug safety and tolerability.
In this study, a final analysis of the RESET was conducted. Patients receiving osimertinib rather than other agents, such as pemetrexed-platinum doublet, as second-line treatments, had longer survival outcomes. Reserving osimertinib for second-line use after failure of first-line afatinib could be a feasible strategy in Asian patients with EGFR+ advanced NSCLC, particularly for those with Del19+. This report suggests that using first- or second-generation TKIs followed by osimertinib could potentially provide a survival benefit in advanced NSCLC patients. However, further prospective trials are required to confirm this strategy and determine the best approach to improving survival outcomes, quality of life, and tolerability in NSCLC patients receiving TKIs.

Electronic Supplementary Material

Supplementary materials are available at Cancer Research and Treatment website (https://www.e-crt.org).

Notes

Ethical Statement

The study for the updated analysis was approved by the Institutional Review Board of the Kosin University Gospel Hospital (KUGH no. 2022-06-038). The study was conducted following the Declaration of Helsinki. All procedures were performed in accordance with relevant guidelines and regulations. The need for informed consent was waived as this study was in retrospective nature.

Author Contributions

Conceived and designed the analysis: Kim T, Jang TW.

Collected the data: Jang TW, Choi CM, Kim MH, Lee SY (Sung Yong Lee), Chang YS, Lee KY, Kim SJ, Yang SH, Ryu JS, Lee JE, Lee SY (Shin Yup Lee), Park CK, Lee SH, Jang SH, Yoon SH, Oh HJ.

Contributed data or analysis tools: Kim T.

Performed the analysis: Kim T.

Wrote the paper: Kim T.

Conflicts of Interest

Conflict of interest relevant to this article was not reported.

Fig. 1
Patient selection process. AJCC, American Joint Committee on Cancer; EGFR, epidermal growth factor receptor; NSCLC, non–small cell lung cancer.
crt-2023-493f1.jpg
Fig. 2
Overall survival between osimertinib and other treatments groups.
crt-2023-493f2.jpg
Fig. 3
Overall survival between osimertinib and pemetrexed-containing agents.
crt-2023-493f3.jpg
Fig. 4
Overall survival comparing osimertinib, pemetrexed-platinum doublet, and pemetrexed monotherapy.
crt-2023-493f4.jpg
Table 1
Baseline characteristics
First-line Afatinib (n=401) Second-line
Osimertinib (n=166) Other therapies (n=235) p-value
Male sex 220 (54.9) 91 (54.8) 129 (54.9) 0.988
Age (yr)
 < 65 221 (55.1) 97 (58.4) 124 (52.8) 0.261
 ≥ 65 180 (44.9) 69 (41.6) 111 (47.2)
Height (cm) 161.5 (8.7) 163.0 (8.6) 160.2 (8.6) 0.014
Weight (kg) 62.3 (11.0) 62.9 (11.8) 61.7 (10.3) 0.411
ECOG PS
 0 and 1 340 (93.2) 135 (95.1) 205 (91.9) 0.292
 ≥ 2 25 (6.8) 7 (4.9) 18 (8.1)
Smoking
 Never 248 (62.5) 101 (61.2) 147 (63.4) 0.902
 Former 111 (28.0) 48 (29.1) 63 (27.2)
 Current 38 (9.6) 16 (9.7) 22 (9.5)
Stagea)
 3 and 4A 234 (58.4) 83 (50.0) 151 (64.3) 0.004
 4B 167 (41.6) 83 (50.0) 84 (35.7)
T category
 T1 60 (27.6) 32 (29.6) 28 (25.7) 0.898
 T2 83 (38.2) 41 (38.0) 42 (38.5)
 T3 24 (11.1) 12 (11.1) 12 (11.0)
 T4 50 (23.0) 23 (21.3) 27 (24.8)
Longest tumor diameter (cm) 3.9 (1.9) 3.8 (2.0) 3.9 (1.8) 0.836
N category
 N0 53 (24.1) 23 (21.1) 30 (27.0) 0.684
 N1 26 (11.8) 13 (11.9) 13 (11.7)
 N2 40 (18.2) 19 (17.4) 21 (18.9)
 N3 101 (45.9) 54 (49.5) 47 (42.3)
M category
 M0 22 (9.9) 9 (8.3) 13 (11.4) 0.345
 M1a 85 (38.1) 37 (33.9) 48 (42.1)
 M1b 38 (17.0) 19 (17.4) 19 (16.7)
 M1c 78 (35.0) 44 (40.4) 34 (29.8)
EGFR mutation
 Del19 222 (55.6) 98 (59.0) 124 (53.2) 0.019
 L858R 123 (30.8) 55 (33.1) 68 (29.2)
 Othersb) 54 (13.5) 13 (7.8) 41 (17.6)
Tissue type
 Adenocarcinoma 394 (98.3) 3 (1.8) 4 (1.7) > 0.99
 Others 7 (1.7) 163 (98.2) 231 (98.3)
No. of metastatic organs
 0–1 202 (50.4) 78 (47.0) 124 (52.8) 0.280
 2–3 167 (41.6) 71 (42.8) 96 (40.9)
 4 or more 32 (8.0) 17 (10.2) 15 (6.4)
Presence of metastasis
 Brain 156 (38.9) 59 (35.5) 97 (41.3) 0.246
 Adrenal gland 32 (8.0) 14 (8.4) 18 (7.7) 0.778
 Lung to lung 127 (31.7) 54 (32.5) 73 (31.1) 0.756
 Liver 50 (12.5) 26 (15.7) 24 (10.2) 0.104
 Bone 168 (41.9) 79 (47.6) 89 (37.9) 0.052
 Pericardial 18 (4.5) 9 (5.4) 9 (3.8) 0.448
 Pleural 158 (39.4) 69 (41.6) 89 (37.9) 0.456
Type of brain metastasis
 Single parenchymal 23 (15.0) 7 (11.7) 16 (17.2) 0.349
 Multiple +/− seeding 130 (85.0) 53 (88.3) 77 (82.8)
New lesion or aggravation of brain metastasis
 No 87 (21.9) 27 (16.3) 60 (25.9) 0.022
 Yes 311 (78.1) 139 (83.7) 172 (74.1)
Dose adjustment for afatinib
 No 151 (37.8) 61 (36.7) 90 (38.5) 0.727
 Yes 249 (62.3) 105 (63.3) 144 (61.5)

Values are presented as number (%). AJCC, American Joint Committee on Cancer; Del19, deletion 19; ECOG PS, Eastern Cooperative Oncology Group performance status; EGFR, epidermal growth factor receptor.

a) Tumor stage was evaluated based on the 8th edition of the AJCC staging manual,

b) Other types of EGFR mutation include compound and uncommon mutations.

Table 2
Time-on-treatment (months) according to the treatments
First-line Afatinib (n=401)
Second-line
Osimertinib (n=166)
Other therapies (n=235)
Median 95% CI p-value Median 95% CI p-value Median 95% CI p-value
Overall 15.0 14.0–16.1 11.9 8.9–14.6 5.1 4.2–5.9
Sex
 Male 14.1 12.6–15.6 0.353 11.8 8.9–14.6 0.603 5.0 4.1–5.8 0.298
 Female 15.8 13.7–17.9 12.4 10.2–14.6 5.6 3.8–7.3
Age (yr)
 < 65 15.7 14.0–17.3 0.162 11.9 9.2–14.7 0.908 5.1 3.8–6.3 0.573
 ≥ 65 14.1 12.3–15.8 11.7 9.4–14.1 5.2 4.0–6.3
BMI (kg/m2)
 < 23.0 17.6 15.9–19.4 0.409 13.5 2.7–24.2 0.815 6.4 4.4–8.4 0.353
 23.0 to < 25.0 17.5 15.4–19.5 14.0 9.2–18.8 7.2 3.3–13.6
 ≥ 25.0 19.1 14.3–23.9 18.1 12.4–23.8 7.3 3.1–13.4
ECOG PS
 0 or 1 15.2 14.0–16.4 0.044 11.8 9.8–13.7 0.869 5.1 4.1–6.0 0.585
 ≥ 2 10.8 6.6–15.0 18.4 10.2–26.6 2.7 0.0–6.7
Smoking
 Never 15.2 13.6–16.9 0.481 12.4 9.9–14.9 0.677 5.4 4.3–6.5 0.718
 Former 14.1 11.1–17.0 11.9 6.9–17.0 4.6 3.5–5.8
 Current 13.9 12.2–15.5 13.5 1.6–25.3 5.1 2.2–8.0
Stagea)
 3 and 4A 16.0 14.5–17.5 < 0.001 13.4 10.4–16.4 0.479 5.7 4.5–7.0 0.183
 4B 13.6 12.7–14.5 11.5 8.8–14.1 4.9 3.8–6.1
T category
 T1 18.2 16.2–20.1 0.019 20.7 9.0–32.4 0.101 8.5 2.0–15.0 0.004
 T2 20.7 17.6–23.8 17.4 11.8–22.9 8.9 5.9–11.8
 T3 15.4 13.2–17.6 10.8 6.3–15.2 9.7 4.7–14.7
 T4 15.7 12.2–19.1 13.7 3.8–23.7 4.7 3.6–5.8
N category
 N0 19.1 14.3–23.9 0.039 20.1 16.3–23.9 0.164 13.3 7.2–19.4 0.007
 N1 23.5 20.2–26.8 10.8 1.4–10.1 5.0 3.7–6.2
 N2 16.2 12.3–20.2 29.1 5.4–52.9 5.2 2.5–7.8
 N3 16.9 14.1–19.7 14.0 7.2–20.8 6.4 4.1–8.7
M category
 M0 18.8 8.8–28.8 0.267 8.8 1.0–16.6 0.570 11.9 6.4–17.4 0.076
 M1a 18.6 17.0–20.2 20.1 11.4–28.8 7.0 3.5–10.4
 M1b 17.5 11.6–23.4 16.1 12.1–20.0 10.2 3.9–16.4
 M1c 16.6 13.8–19.3 11.8 3.7–19.8 5.2 3.7–6.6
Tissue type
 Adenocarcinoma 15.2 14.0–16.4 0.003 12.4 10.6–14.2 0.082 5.1 4.3–5.9 0.318
 Others 10.3 4.6–15.9 9.4 0.0–18.8 3.0 4.3–5.9
EGFR
 Del19 15.7 14.1–17.3 0.037 13.0 8.2–17.8 0.755 5.0 3.6–6.3 0.297
 L858R 15.4 13.1–17.8 11.2 8.6–13.8 5.4 4.5–6.3
 Othersb) 11.7 9.9–13.5 13.4 6.9–20.0 5.2 1.9–8.6
No. of metastatic organs
 0–1 16.6 15.0–18.2 < 0.001 13.7 10.3–17.2 0.311 6.5 4.7–8.2 < 0.001
 2–3 13.9 12.4–15.4 11.8 9.4–14.1 4.8 3.8–5.9
 4 or more 11.2 8.6–13.9 8.7 7.4–10.0 2.2 1.8–2.5
Brain metastasis
 No 15.9 14.4–17.4 0.037 11.5 9.4–13.6 0.757 5.4 4.2–6.6 0.478
 Yes 13.4 12.3–14.5 13.0 9.7–16.3 5.0 3.9–6.0
Adrenal gland metastasis
 No 15.2 14.1–16.3 0.945 11.5 9.6–13.4 0.467 5.1 4.3–5.9 0.447
 Yes 13.3 10.1–16.4 14.5 3.1–25.8 5.8 0.2–11.5
Lung to lung metastasis
 No 15.0 13.7–16.3 0.542 11.5 9.4–13.6 0.219 5.9 4.9–7.0 0.070
 Yes 14.9 12.5–17.4 13.0 5.6–20.5 4.2 3.0–5.4
Liver metastasis
 No 15.8 14.3–17.3 < 0.001 14.0 9.2–18.8 < 0.001 5.5 4.6–6.4 < 0.001
 Yes 10.3 7.5–13.0 8.5 6.1–10.9 2.1 1.3–2.9
Bone metastasis
 No 15.9 14.4–17.4 0.026 14.0 9.0–19.0 0.194 5.9 4.5–7.4 0.013
 Yes 13.9 13.0–14.8 11.3 9.4–13.2 4.4 3.4–5.4
Pericardial metastasis
 No 15.2 14.0–16.4 0.258 12.4 10.6–14.2 0.216 5.2 4.4–6.0 0.006
 Yes 9.4 5.3–13.6 8.1 5.7–10.4 2.7 2.4–3.0
Pleural metastasis
 No 14.7 13.4–16.0 0.982 12.6 9.3–15.9 0.407 5.4 4.2–6.5 0.210
 Yes 15.2 12.9–17.5 11.9 5.7–18.2 4.4 3.4–5.4
Type of brain metastasis
 Single parenchymal 14.8 8.8–20.8 0.534 17.5 17.1–17.8 0.171 3.0 0.0–8.9 0.757
 Multiple +/− seeding 13.3 12.1–14.4 11.9 8.3–15.6 5.0 4.1–5.9
New lesion or aggravation of brain metastasis
 No 15.4 13.9–16.9 0.137 12.4 10.0–14.8 0.280 6.2 4.9–7.4 0.003
 Yes 12.7 10.6–14.9 11.3 6.1–16.4 3.4 2.2–4.7
Dose adjustment for afatinib
 No 12.9 11.7–14.1 0.002 13.0 9.8–16.1 0.892 5.0 3.9–6.0 0.228
 Yes 16.6 15.3–17.9 11.9 9.4–14.4 5.6 4.2–6.9

AJCC, American Joint Committee on Cancer; BMI, body mass index; CI, confidence interval; Del19, deletion 19; ECOG PS, Eastern Cooperative Oncology Group performance status; EGFR, epidermal growth factor receptor.

a) Tumor stage was evaluated based on the 8th edition of the AJCC staging manual,

b) Other types of EGFR mutation include compound and uncommon mutations.

Table 3
Overall survival (months) according to the treatments
1′ Afatinib → 2′ Osimertinib (n=166)
1′ Afatinib → 2′ Other therapies (n=235)
Median 95% CI p-value Median 95% CI p-value
Overall 54.3 46.7–61.9 41.3 32.9–49.8
Sex
 Male 61.4 50.2–72.6 0.251 36.6 28.0–45.3 0.079
 Female 51.5 44.1–58.8 41.7 30.2–53.2
Age (yr)
 < 65 52.8 40.3–65.2 0.541 41.7 21.7–61.7 0.618
 ≥ 65 58.9 50.7–67.2 41.3 32.4–50.2
BMI (kg/m2)
 < 23.0 61.4 46.8–76.0 0.238 66.6 62.9–70.3 0.845
 23.0 to < 25.0 62.9 46.2–79.7 NR 41.7-NA
 ≥ 25.0 NR 52.8-NA NR 41.7-NA
ECOG PS
 0 or 1 59.1 48.5–69.8 0.012 41.7 31.1–52.3 0.321
 ≥ 2 29.5 14.6–44.6 33.6 16.9–50.4
Smoking
 Never 58.9 51.6–66.3 0.293 41.1 31.1–51.2 0.724
 Former 49.1 42.4–55.8 41.7 29.4–55.0
 Current NR 47.7-NA 29.1 14.8-NA
Stagea)
 3 and 4A 62.9 51.5-NA 0.020 50.0 33.2–66.9 0.027
 4B 48.5 44.0-NA 34.0 25.0–43.0
T category
 T1 NR 61.4-NA 0.002 NR 60.6-NA 0.110
 T2 NR 61.7-NA NR 49.0-NA
 T3 62.9 38.3-NA NR 24.2-NA
 T4 51.1 45.2-NA 65.1 25.0–105.2
N category
 N0 NR 52.8-NA 0.596 NR NA-NA 0.016
 N1 59.1 44.1–74.1 NR NA-NA
 N2 NR 62.9-NA NR NA-NA
 N3 61.4 49.1–73.7 60.6 29.1–92.0
M category
 M0 NR 42.2-NA 0.375 NR NA-NA 0.032
 M1a NR 54.3-NA NR NA-NA
 M1b 61.4 NA-NA 39.4 35.1–43.6
 M1c 58.9 41.9–76.0 66.6 31.9–101.3
Tissue type
 Adenocarcinoma 54.3 45.9–61.4 0.050 41.7 32.8–50.5 0.154
 Others 44.0 20.0-NA 9.4 6.6-NA
EGFR
 Del19 59.1 48.7–69.5 0.422 65.1 40.5–89.7 0.051
 L858R 46.5 33.2–59.7 41.3 36.9–45.8
 Othersb) 45.2 19.7–70.8 31.9 26.8–37.0
No. of metastatic organs
 0–1 62.9 52.8-NA 0.003 60.6 42.3–79.0 < 0.001
 2–3 46.6 40.9–52.2 35.3 25.0–45.7
 4 or more 28.7 19.6–37.8 25.6 17.7–33.4
Brain metastasis
 No 59.1 49.6–68.7 0.044 47.8 32.2–63.4 0.215
 Yes 48.5 40.6–56.4 37.2 28.7–45.7
Adrenal gland metastasis
 No 52.8 43.2–62.3 0.190 41.7 31.2–52.2 0.186
 Yes NR 48.5-NA 35.5 23.1–48.0
Lung to lung metastasis
 No 52.8 43.4–62.1 0.815 41.7 31.4–51.9 0.889
 Yes 54.3 39.8–68.8 39.4 16.3–62.4
Liver metastasis
 No 61.4 64.0–68.8 < 0.001 41.7 31.5–51.8 0.001
 Yes 31.9 20.5–43.4 23.7 18.3–29.0
Bone metastasis
 No 59.1 51.5-NA 0.027 52.9 34.2–71.6 0.002
 Yes 48.5 36.6–60.4 30.2 23.2–37.2
Pericardial metastasis
 No 58.9 50.2–67.6 0.053 41.7 31.9–51.5 0.026
 Yes 46.6 0.0–99.9 25.6 22.3–28.8
Pleural metastasis
 No 54.3 39.6–69.0 0.645 39.4 28.0–50.7 0.810
 Yes 52.5 40.3–64.7 41.7 31.8–51.6
Type of brain metastasis
 Single parenchymal 49.1 15.4-NA 0.810 37.3 18.1–56.4 0.944
 Multiple +/‒ seeding 46.5 31.8–61.2 36.9 26.7–47.2
New lesion or aggravation of brain metastasis
 No 59.1 49.5–68.8 0.011 41.7 25.2–58.2 0.342
 Yes 30.9 10.5–51.4 38.8 29.6–47.9
Dose adjustment for afatinib
 No 62.9 44.7–81.2 0.302 34.2 23.4–45.0 0.026
 Yes 52.5 41.4–63.7 50.0 32.7–67.4

AJCC, American Joint Committee on Cancer; BMI, body mass index; CI, confidence interval; Del19, deletion 19; ECOG PS, Eastern Cooperative Oncology Group performance status; EGFR, epidermal growth factor receptor; NA, not available.

a) Tumor stage was evaluated based on the 8th edition of the AJCC staging manual,

b) Other types of EGFR mutation include compound and uncommon mutations.

Table 4
Factors affecting time-on-treatment during first-line afatinib
Univariable
Multivariable
HR (95% CI) p-value HR (95% CI) p-value
Sex
 Male 1 -
 Female 1.10 (0.89–1.36) 0.354
Age (yr)
 < 65 1 -
 ≥ 65 1.16 (0.94–1.43) 0.163
BMI (kg/m2)
 < 23.0 1 -
 23.0 to < 25.0 1.18 (0.82–1.70) 0.381
 ≥ 25.0 0.56 (0.66–1.26) 0.563
ECOG PS
 0 or 1 1 1
 ≥ 2 1.54 (1.01–2.36) 0.046 1.62 (1.04–2.53) 0.033
Smoking
 Never 1 -
 Former 1.16 (0.91–1.47) 0.230
 Current 1.02 (0.71–1.47) 0.898
Stagea)
 3 and 4A 1 1
 4B 1.49 (1.20–1.84) < 0.001 1.16 (0.86–1.56) 0.321
T category
 T1 1 -
 T2 0.76 (0.53–1.08) 0.129
 T3 1.48 (0.89–2.47) 0.133
 T4 1.23 (0.82–1.84) 0.313
N category
 N0 1 -
 N1 0.98 (0.60–1.60) 0.944
 N2 1.35 (0.86–2.10) 0.191
 N3 1.58 (1.10–2.27) 0.013
M category
 M0 1 -
 M1a 1.10 (0.65–1.86) 0.715
 M1b 1.31 (0.73–2.36) 0.372
 M1c 1.48 (0.87–2.52) 0.149
Tissue type
 Adenocarcinoma 1 1
 Others 2.98 (1.40–6.35) 0.005 4.25 (1.85–9.76) < 0.001
EGFR
 Del19 1 1
 L858R 1.12 (0.88–1.41) 0.362 1.12 (0.86–1.44) 0.401
 Othersb) 1.50 (1.10–2.05) 0.011 1.57 (1.13–2.18) 0.007
No. of metastatic organs
 0–1 1 1
 2–3 1.48 (1.18–1.85) 0.001 1.63 (1.17–2.26) 0.004
 4 or more 1.94 (1.32–2.86) 0.001 2.44 (1.38–4.32) 0.002
Brain metastasis
 No 1 1
 Yes 1.26 (1.01–1.56) 0.038 0.98 (0.75–1.27) 0.858
Adrenal gland metastasis
 No 1 -
 Yes 1.01 (0.69–1.49) 0.945
Lung to lung metastasis
 No 1 -
 Yes 1.07 (0.86–1.34) 0.543
Liver metastasis
 No 1 1
 Yes 1.88 (1.36–2.58) < 0.001 1.22 (0.81–1.81) 0.349
Bone metastasis
 No 1 1
 Yes 1.27 (1.03–1.57) 0.027 0.8 (0.59–1.08) 0.141
Pericardial metastasis
 No 1 -
 Yes 1.31 (0.82–2.11) 0.260
Pleural metastasis
 No 1 -
 Yes 1.00 (0.81–1.24) 0.982
Type of brain metastasis
 Single parenchymal 1 -
 Multiple +/‒ seeding 1.16 (0.73–1.82) 0.535
New lesion or aggravation of brain metastasis
 No 1 -
 Yes 1.21 (0.94–1.55) 0.138
Dose adjustment for afatinib
 Yes 1 1
 No 1.42 (1.14–1.76) 0.002 1.63 (1.29–2.07) 0.005

AJCC, American Joint Committee on Cancer; BMI, body mass index; CI, confidence interval; Del19, deletion 19; ECOG PS, Eastern Cooperative Oncology Group performance status; EGFR, epidermal growth factor receptor; HR, hazard ratio; meta, metastasis; PS, performance status.

a) Tumor stage was evaluated based on the 8th edition of the AJCC staging manual,

b) Other types of EGFR mutation include compound and uncommon mutations.

Table 5
Factors affecting overall survival during first- and second-line treatments
1′ Afatinib → 2′ Osimertinib (n=166)
1′ Afatinib → 2′ Other therapies (n=235)
Univariable
Multivariable
Univariable
Multivariable
HR (95% CI) p-value HR (95% CI) p-value HR (95% CI) p-value HR (95% CI) p-value
Sex
 Male 1 - 1 1
 Female 1.33 (0.81–2.19) 0.253 0.70 (0.47–1.04) 0.080 0.76 (0.50–1.17) 0.210
Age (yr)
 < 65 1 - 1 -
 ≥ 65 0.85 (0.51–1.43) 0.541 1.11 (0.74–1.64) 0.618
BMI (kg/m2)
 < 23.0 1 - 1 -
 23.0 to < 25.0 0.37 (0.11–1.28) 0.115 1.27 (0.55–2.91) 0.573
 ≥ 25.0 0.70 (0.33–1.52) 0.369 1.05 (0.49–2.24) 0.909
ECOG PS
 0 or 1 1 1 1 -
 ≥ 2 3.1 (1.22–7.83) 0.017 2.79 (1.01–7.71) 0.047 1.44 (0.70–2.98) 0.324
Smoking
 Never 1 - 1 -
 Former 1.23 (0.72–2.10) 0.457 0.95 (0.60–1.52) 0.838
 Current 0.57 (0.22–1.46) 0.244 1.30 (0.65–2.60) 0.468
Stagea)
 3 and 4A 1 1 1 1
 4B 1.61 (0.97–2.68) 0.065 0.71 (0.30–1.72) 0.450 1.58 (1.05–2.38) 0.028 1.19 (0.72–1.97) 0.493
T category
 T1 1 - 1 -
 T2 1.08 (0.39–2.97) 0.887 1.35 (0.53–3.43) 0.529
 T3 1.65 (0.43–6.39) 0.469 2.43 (0.70–8.37) 0.161
 T4 3.49 (1.37–8.97) 0.009 2.68 (1.06–6.73) 0.036
N category
 N0 1 - 1 -
 N1 0.89 (0.23–3.47) 0.872 0.89 (0.17–4.62) 0.890
 N2 0.75 (0.23–2.41) 0.626 2.89 (0.93–8.99) 0.066
 N3 1.44 (0.58–3.54) 0.433 3.65 (1.36–9.81) 0.010
M category
 M0 1 - 1 -
 M1a 1.24 (0.28–5.61) 0.777 1.84 (0.41–8.22) 0.426
 M1b 1.08 (0.19–5.98) 0.928 4.29 (0.80–20.39) 0.067
 M1c 2.21 (0.49–9.89) 0.298 4.38 (0.99–19.39) 0.051
Tissue type
 Adenocarcinoma 1 - 1 -
 Others 3.09 (0.75–12.75) 0.120 2.68 (0.65–10.96) 0.171
EGFR
 Del19 1 - 1 1
 L858R 1.37 (0.80–2.32) 0.249 1.27 (0.81–2.00) 0.293 1.41 (0.89–2.22) 0.146
 Othersb) 1.45 (0.64–3.28) 0.371 1.90 (1.12–3.21) 0.017 1.88 (1.08–3.28) 0.026
No. of metastatic organs
 0–1 1 1 1
 2–3 1.86 (1.08–3.21) 0.025 1.15 (0.49–2.72) 0.750 2.12 (1.40–3.21) < 0.001 1.57 (0.88–2.79) 0.128
 4 or more 3.26 (1.55–6.87) 0.002 1.74 (0.49–6.21) 0.395 3.27 (1.45–7.39) 0.004 2.03 (0.68–6.08) 0.204
Brain metastasis
 No 1 1 1 -
 Yes 1.68 (1.01–2.80) 0.047 1.75 (0.86–3.58) 0.122 1.29 (0.86–1.91) 0.217
Adrenal gland metastasis
 No 1 - 1 -
 Yes 0.47 (0.15–1.49) 0.201 1.63 (0.79–3.37) 0.190
Lung to lung metastasis
 No 1 - 1 -
 Yes 1.06 (0.64–1.77) 0.815 1.03 (0.68–1.57) 0.889
Liver metastasis
 No 1 1 1 1
 Yes 3.69 (2.03–6.72) < 0.001 2.04 (0.92–4.57) 0.081 2.62 (1.42–4.82) 0.002 1.89 (0.92–3.89) 0.084
Bone metastasis
 No 1 1 1 1
 Yes 1.74 (1.06–2.86) 0.029 1.19 (0.51–2.77) 0.686 1.87 (1.25–2.81) 0.002 1.20 (0.69–2.07) 0.519
Pericardial metastasis
 No 1 1 1 1
 Yes 2.26 (0.97–5.28) 0.060 2.08 (0.69–6.34) 0.195 2.49 (1.08–5.72) 0.032 1.42 (0.56–3.60) 0.464
Pleural metastasis
 No 1 - 1 -
 Yes 0.89 (0.54–1.47) 0.646 1.05 (0.71–1.57) 0.810
Type of brain metastasis
 Single parenchymal 1 - 1 -
 Multiple +/− seeding 1.16 (0.34–3.92) 0.810 0.97 (0.46–2.06) 0.944
New lesion or aggravation of brain metastasis
 No 1 1 1 -
 Yes 2.05 (1.16–3.61) 0.013 1.68 (0.83–3.40) 0.146 1.24 (0.79–1.95) 0.343
Dose adjustment for afatinib
 Yes 1 - 1 1
 No 0.75 (0.44–1.29) 0.303 1.56 (1.05–2.32) 0.027 1.61 (1.05–2.48) 0.029

AJCC, American Joint Committee on Cancer; BMI, body mass index; CI, confidence interval; Del19, deletion 19; ECOG PS, Eastern Cooperative Oncology Group performance status; EGFR, epidermal growth factor receptor; HR, hazard ratio.

a) Tumor stage was evaluated based on the 8th edition of the AJCC staging manual,

b) Other types of EGFR mutation include compound and uncommon mutations.

Table 6
Comparison of RESET with other previous real-world studies
Asian Median OS (95% CI, mo)
RESET 153 54.3 (46.7–61.9)
 Del19 98 59.1 (48.7–69.5)
 L858R 55 46.5 (33.2–59.7)
GioTag+UpSwinG [15] 168 45.2 (41.7–71.1)
 Del19 109 63.5 (42.3–71.1)
 L858R 59 39.1 (29.3–48.5)

CI, confidence interval; Del19, deletion 19; OS, overall survival.

References

1. Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Pineros M, Znaor A, et al. Cancer statistics for the year 2020: an overview. Int J Cancer. 2021;149:778–89.
crossref pdf
2. Zhang Y, Luo G, Etxeberria J, Hao Y. Global patterns and trends in lung cancer incidence: a population-based study. J Thorac Oncol. 2021;16:933–44.
crossref pmid
3. Cheng TY, Cramb SM, Baade PD, Youlden DR, Nwogu C, Reid ME. The international epidemiology of lung cancer: latest trends, disparities, and tumor characteristics. J Thorac Oncol. 2016;11:1653–71.
crossref pmid pmc
4. Lortet-Tieulent J, Soerjomataram I, Ferlay J, Rutherford M, Weiderpass E, Bray F. International trends in lung cancer incidence by histological subtype: adenocarcinoma stabilizing in men but still increasing in women. Lung Cancer. 2014;84:13–22.
crossref pmid
5. Lee JG, Kim HC, Choi CM. Recent trends of lung cancer in Korea. Tuberc Respir Dis. 2021;84:89–95.
crossref pmid pmc pdf
6. Soria JC, 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.
crossref pmid
7. Ke EE, Zhou Q, Zhang QY, Su J, Chen ZH, Zhang XC, et al. A higher proportion of the EGFR T790M mutation may contribute to the better survival of patients with exon 19 deletions compared with those with L858R. J Thorac Oncol. 2017;12:1368–75.
crossref pmid
8. Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med. 2017;376:629–40.
crossref pmid pmc
9. Paz-Ares L, Tan EH, O’Byrne K, Zhang L, Hirsh V, Boyer M, et al. Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: overall survival data from the phase IIb LUX-Lung 7 trial. Ann Oncol. 2017;28:270–7.
crossref pmid pmc
10. Hochmair MJ, Morabito A, Hao D, Yang CT, Soo RA, Yang JC, et al. Sequential afatinib and osimertinib in patients with EGFR mutation-positive non-small-cell lung cancer: final analysis of the GioTag study. Future Oncol. 2020;16:2799–808.
crossref pmid
11. Popat S, Jung HA, Lee SY, Hochmair MJ, Lee SH, Escriu C, et al. Sequential afatinib and osimertinib in patients with EGFR mutation-positive NSCLC and acquired T790M: a global non-interventional study (UpSwinG). Lung Cancer. 2021;162:9–15.
crossref pmid
12. Kim T, Jang TW, Choi CM, Kim MH, Lee SY, Park CK, et al. Sequential treatment of afatinib and osimertinib or other regimens in patients with advanced non-small-cell lung cancer harboring EGFR mutations: results from a real-world study in South Korea. Cancer Med. 2021;10:5809–22.
crossref pmid pmc pdf
13. Wu YL, Zhou C, Hu CP, Feng J, Lu S, Huang Y, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:213–22.
crossref pmid
14. Akamatsu H, Katakami N, Okamoto I, Kato T, Kim YH, Imamura F, et al. Osimertinib in Japanese patients with EGFR T790M mutation-positive advanced non-small-cell lung cancer: AURA3 trial. Cancer Sci. 2018;109:1930–8.
pmid pmc
15. Smit EF, Burgers SA, Biesma B, Smit HJ, Eppinga P, Dingemans AM, et al. Randomized phase II and pharmacogenetic study of pemetrexed compared with pemetrexed plus carboplatin in pretreated patients with advanced non-small-cell lung cancer. J Clin Oncol. 2009;27:2038–45.
crossref pmid
16. Miura S, Jung HA, Lee SY, Lee SH, Lee MK, Lee YC, et al. Sequential afatinib and osimertinib in Asian patients with EGFR mutation-positive non-dmall vell lung cancer and acquired T790M: combined analysis of two global non-interventional studies. Onco Targets Ther. 2022;15:873–82.
pmid pmc
17. Kogure Y, Shigematsu F, Oki M, Saka H. T790M correlates with longer progression-free survival in non-small cell lung carcinomas harboring EGFR mutations. In Vivo. 2018;32:1199–204.
crossref pmid pmc
18. Gaut D, Sim MS, Yue Y, Wolf BR, Abarca PA, Carroll JM, et al. Clinical implications of the T790M mutation in disease characteristics and treatment response in patients with epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer (NSCLC). Clin Lung Cancer. 2018;19:e19–28.
crossref pmid pmc
19. Chmielecki J, Foo J, Oxnard GR, Hutchinson K, Ohashi K, Somwar R, et al. Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling. Sci Transl Med. 2011;3:90ra59.
crossref pmid pmc
20. Regales L, Balak MN, Gong Y, Politi K, Sawai A, Le C, et al. Development of new mouse lung tumor models expressing EGFR T790M mutants associated with clinical resistance to kinase inhibitors. PLoS One. 2007;2:e810.
crossref pmid pmc
21. Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E, Tiseo M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer. 2019;121:725–37.
crossref pmid pmc pdf
22. Yonesaka K, Kobayashi Y, Hayashi H, Chiba Y, Mitsudomi T, Nakagawa K. Dual blockade of EGFR tyrosine kinase using osimertinib and afatinib eradicates EGFR-mutant Ba/F3 cells. Oncol Rep. 2019;41:1059–66.
crossref pmid
23. Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med. 2020;382:41–50.
crossref pmid
24. Kang MJ, Won YJ, Lee JJ, Jung KW, Kim HJ, Kong HJ, et al. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2019. Cancer Res Treat. 2022;54:330–44.
crossref pmid pmc pdf
Editorial Office
Korean Cancer Association
Room 1824, Gwanghwamun Officia
92 Saemunan-ro, Jongno-gu, Seoul 03186, Korea
TEL: +82-2-3276-2410   FAX: +82-2-792-1410   E-mail: journal@cancer.or.kr
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Copyright © Korean Cancer Association.                 Developed in M2PI