EGFR inhibitors plus dabrafenib and trametinib in patients with EGFR-mutant lung cancer and resistance mediated by BRAFV600E mutation: a multi-center real-world experience in China
Original Article

EGFR inhibitors plus dabrafenib and trametinib in patients with EGFR-mutant lung cancer and resistance mediated by BRAFV600E mutation: a multi-center real-world experience in China

Wenyue Yang1,2,3# ORCID logo, Xiangran Feng4# ORCID logo, Jian Ni5, Xin Zhang1,2,3 ORCID logo, Hui Yu1,2,3, Xianghua Wu1,2,3, Huijie Wang1,2,3, Xinmin Zhao1,2,3, Zhihuang Hu1,2,3, Bo Yu1,2,3, Yao Zhang1,2,3, Ying Lin1,2,3, Yi Xiang4 ORCID logo, Jialei Wang1,2,3 ORCID logo

1Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; 2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; 3Institute of Thoracic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; 4Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai, China; 5Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China

Contributions: (I) Conception and design: Y Xiang, J Wang; (II) Administrative support: Y Xiang, J Wang, H Yu, X Wu, H Wang, X Zhao, Z Hu; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: W Yang, X Feng, J Ni, X Zhang, B Yu, Y Zhang, Y Lin; (V) Data analysis and interpretation: W Yang, X Feng; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work as co-first authors.

Correspondence to: Jialei Wang, MD. Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270 Dong’an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Thoracic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. Email: wangjialei@shca.org.cn; Yi Xiang, MD, PhD. Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, No. 197, Ruijin 2nd Road, Shanghai 200025, China. Email: xiangyiht@163.com.

Background: The combination therapy of the B-Raf proto-oncogene (BRAF) inhibitor dabrafenib and the mitogen-activated protein kinase kinase (MEK) inhibitor Trametinib has shown favorable outcomes in patients initially identified with BRAFV600E mutations. However, there are currently no large-scale study data focusing on the use of a triple therapy regimen of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) plus dabrafenib and trametinib in patients with newly concomitant BRAF mutations after acquiring resistance to EGFR-TKIs. Our study aimed to explore the efficacy and safety of the triple therapy regimen through a multi-center real-world experience.

Methods: We reviewed the medical records of 1,861 patients who were treated with EGFR-TKI targeted drugs at three major medical centers in Shanghai between June 2015 and August 2024. Among 1,288 patients who developed disease progression, we identified 14 patients who were treated with a triple therapy regimen of EGFR-TKI plus dabrafenib and trametinib due to newly acquired BRAFV600E mutation after EGFR-TKI resistance. The assessments comprised progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), and adverse events (AEs). We also performed further subgroup analysis to aid in identifying potential factors that influence treatment outcomes and enhance clinical decision-making.

Results: At the time of the data cutoff (August 1, 2024), the estimated median PFS was 6.7 months [95% confidence interval (CI): 2.5–not evaluated (NE)]. The median OS was not reached in 14 patients. ORR was 35.7% (95% CI: 14.0–64.4%) and DCR was 78.6% (95% CI: 52.4–92.4%). Three patients (21.4%) reported progressive disease (PD) and that was the best response. The median PFS was 8.35 months (95% CI: 2.0–NE) in 8 patients receiving third-generation TKI followed by first-/second-generation EGFR-TKIs and 6.9 months (95% CI: 2.5–NE) in 6 patients receiving third-generation TKI as first-line treatment directly. There was no significant difference in PFS between the two groups of patients receiving third-generation TKIs in different treatment sequences above [hazard ratio (HR): 1.107; 95% CI: 0.318–3.854; P=0.85]. Subgroup analysis indicated that a complex genetic mutation background may be a potential factor contributing to poorer PFS. No unexpected adverse effects were reported. Apart from pyrexia, gastrointestinal-related adverse reactions and skin-related adverse reactions warrant close attention.

Conclusions: The triple therapy regimen of EGFR-TKI plus dabrafenib and trametinib was found to have substantial and durable clinical benefit, with a manageable safety profile, in patients with newly concomitant BRAFV600E mutations after osimertinib failure.

Keywords: B-Raf proto-oncogene (BRAF); epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs); acquired resistance; non-small cell lung cancer (NSCLC)


Submitted Sep 04, 2024. Accepted for publication Nov 26, 2024. Published online Dec 27, 2024.

doi: 10.21037/tlcr-24-803


Highlight box

Key findings

• The triple regimen of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) plus dabrafenib and trametinib was found to have substantial clinical benefit in patients with newly concomitant BRAFV600E mutations after osimertinib failure.

• The triple regimen did not lead to fatal adverse reactions and the safety profile was considered manageable.

What is known and what is new?

• Treatment failure of EGFR-TKIs is a critical challenge that need to be addressed in advanced non-small cell lung cancer, with B-Raf proto-oncogene mutation being considered as one of the resistance mechanisms.

• The significant differences in progression-free survival and response rates between patients with a complex genetic mutation background and those with only BRAFV600E mutation may provide valuable insights for clinical practice.

What is the implication, and what should change now?

• It is worth noting the importance of utilizing next-generation sequencing (NGS) to identify resistance mechanisms following osimertinib failure, as well as the need for high-quality biopsy samples and appropriate timing of testing, since NGS results directly impact patient treatment decisions.


Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide (1). Although significant advances have been made in the treatment of non-small cell lung cancer (NSCLC) over the past 30 years by identifying driver gene mutations and developing targeted therapies, lung cancer remains a significant public health problem due to its high incidence. The best-known oncogenic drivers of NSCLC involve activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene, with the most frequent mutations being the exon 19 deletion and the exon 21 L858R substitution (2). The two most common EGFR alterations represent approximately 85–90% of all EGFR alterations in NSCLC and both result in activation of the tyrosine kinase domain and are associated with sensitivity to small-molecule EGFR tyrosine kinase inhibitors (TKIs) (3). However, whether it is 9.5 months of progression-free survival (PFS) in the IPASS trial, 11.1 months of PFS in the LUX-Lung 3 trial, or 18.9 months of PFS in the FLAURA trial, acquired patient resistance to EGFR-TKIs remains an area that needs further exploration (4-6). The currently known acquired resistance mechanisms to EGFR TKIs can be broadly categorized into on-target mechanisms, such as the EGFRT790M/EGFRC797S mutation and EGFR amplification, as well as off-target mechanisms, including Rat sarcoma-mitogen-activated protein kinase (RAS-MAPK) pathway aberrations, bypass signaling activation, and histologic or phenotypic transformation (7-9).

RAS-MAPK pathway aberrations that are known to confer resistance to osimertinib include B-Raf proto-oncogene (BRAF), neuroblastoma RAS viral oncogene homolog (NRAS), and V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations. BRAF mutations are found in 2–5% of NSCLC cases, with approximately 50% being the BRAFV600E variant (10). Although the frequency of this mutation as a secondary targetable alteration in osimertinib-resistant patients is much lower, studies have confirmed that new driver gene alterations in EGFR-mutated lung cancer can serve as a mechanism of resistance to osimertinib (11,12). Non-V600 BRAF mutations, such as G469A, have also been reported as an acquired resistance mechanism to osimertinib (13). Based on the data from the clinical trial BRF113928 (NCT01336634) [dabrafenib and trametinib combination group—objective response rate (ORR): 63%, median progression free survival (mPFS): 10.2 months, median overall survival (mOS): 18.2 months], the combination of BRAF inhibitor dabrafenib and mitogen-activated protein kinase kinase (MEK) inhibitor trametinib was approved by the Food and Drug Administration (FDA) in June, 2017 for the treatment of BRAFV600E mutation-positive NSCLC (14). Despite the excellent efficacy demonstrated by the combination of dabrafenib and trametinib in patients initially identified with BRAFV600E mutations, the absence of data from large-scale clinical studies on patients who develop BRAF mutations following resistance to EGFR-TKIs remains a major obstacle in selecting treatment options for these patients.

In this study, we reviewed the treatment histories of 14 patients with newly acquired BRAFV600E mutation among 1,288 patients who developed EGFR-TKI resistance. Based on the studied population of patients, we estimated the median PFS for the triple therapy regimen of EGFR-TKI plus dabrafenib and trametinib. Our subgroup analysis can also help identify potential factors influencing treatment outcomes and promote clinical decision-making. We present this article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-803/rc).


Methods

Study design and patients enrollment

We reviewed the medical records of 1,861 patients who were treated with EGFR-TKI targeted drugs at three major medical centers in Shanghai between June 2015 and August 2024. Among 1,288 patients who developed disease progression, we identified 14 patients who were diagnosed with NSCLC with either EGFR19del or EGFRL858R mutations and after treatment with 1st/2nd/3rd generation of EGFR-TKI, then blood or tissue-based next-generation sequencing (NGS) was performed to identify BRAFV600E mutation as potential resistance mechanisms. Data were cut-off on August 1, 2024. Patients who did not complete tumor response assessments were excluded from the study.

This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013), and the protocol was reviewed and approved by the institutional review board of Ruijin Hospital (No. 2024-164), the Ethics Committee of Fudan University Shanghai Cancer Center (No. 1612167-18) and the Ethics Committee of Shanghai Pulmonary Hospital (No. L22-407). Individual consent for this retrospective analysis was waived.

Effectiveness and safety evaluation

Effectiveness was assessed by PFS, overall survival (OS), ORR, and disease control rate (DCR). PFS was defined as the time from initiation of dabrafenib plus trametinib therapy to the disease progression or death. OS was defined as the time from using dabrafenib plus trametinib therapy to death due to any cause. DCR was defined as the proportion of patients achieving complete response (CR), partial response (PR) or stable disease (SD). ORR was defined as the proportion of patients achieving CR or PR. Tumor response was initially evaluated after 1 month of treatment with dabrafenib and trametinib, and subsequently every 2 months, based on the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1.

Adverse events (AEs) were monitored monthly in accordance with the National Cancer Institute’s Common Terminology Criteria for Adverse Events (CTCAE), version 5.0.

Survival outcomes and disease progression data were acquired through medical record review and telephone interviews conducted by trained staff members. In cases the patients or their family members were unreachable at the scheduled follow-up date, the date and survival information were censored according to the last follow-up.

Statistical analysis

Categorical variables were summarized by frequency and percentage, while continuous variables were described using medians and ranges. PFS and OS were estimated using the Kaplan-Meier method, accompanied by hazard ratios (HRs). All outcome measures were reported with 95% confidence intervals (CIs), estimated through the log-rank proportional hazards model. Due to the limited sample size and inability to satisfy the proportional hazards assumption of Cox regression, exploratory univariate analyses were conducted using the log-rank test. AEs were summarized by frequency counts and percentages.

All P values were two-sided, with statistical significance set at P<0.05. Analyses were performed using IBM® SPSS® Statistics version 26 and R version 4.0.2, with graphs created in R version 4.0.2.


Results

Patient baseline characteristics

Fourteen patients with newly acquired BRAFV600E mutation after EGFR-TKI resistance at three major medical centers in Shanghai between June 2015 and August 2024 were included in the study (Figure 1). A total of 1,124 patients underwent NGS testing after developing resistance to EGFR-TKIs, revealing a BRAFV600E incidence rate of 1.25% in this setting and the incidence rates in the respective groups were 1.65% and 1.32%. The baseline characteristics of the patients at the start of dabrafenib plus trametinib therapy are outlined in Table 1. The median age of the patients was 60 years (range, 53 to 70 years); 57.1% (8 out of 14) were female, and 64.3% (9 out of 14) were never-smokers. The majority of patients had an Eastern Cooperative Oncology Group (ECOG) score of 1 and were stage IV disease at baseline of dabrafenib plus trametinib therapy; 42.9% (6/14) of patients had a surgical history at the time of initial diagnosis of lung cancer.

Figure 1 Flowchart of the patient selection process. NSCLC, non-small cell lung cancer; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; NGS, next-generation sequencing; D+T + EGFR-TKI, dabrafenib and trametinib plus epidermal growth factor receptor-tyrosine kinase inhibitor.

Table 1

Baseline patient characteristics

Characteristics Value (N=14)
Sex, n (%)
   Male 6 (42.9)
   Female 8 (57.1)
Age (years)
   Median [range] 60 [53–70]
   <60 7 (50.0)
   ≥60 7 (50.0)
History of previous smoking, n (%)
   Yes 5 (35.7)
   No 9 (64.3)
ECOG performance status, n (%)
   1 12 (85.7)
   2 2 (14.3)
Stage, n (%)
   III 3 (21.4)
   IV 11 (78.6)
Surgical history, n (%)
   Yes 6 (42.9)
   No 8 (57.1)
Number of metastatic sites, n (%)
   0 3 (21.4)
   1–2 10 (71.4)
   ≥3 1 (7.2)
Metastatic site, n (%)
   Bone 3 (21.4)
   Contralateral lung 5 (35.7)
   Brain 3 (21.4)
   Pleura 2 (14.3)
   Adrenal gland 1 (7.2)
EGFR mutation, n (%)
   EGFR 19 del 10 (71.4)
   EGFR L858R 3 (21.4)
   EGFR 19 del + EGFR L858R 1 (7.2)
First-line treatment, n (%)
   1st EGFR-TKI 6 (42.9)
   2nd EGFR-TKI 2 (14.2)
   3rd EGFR-TKI 6 (42.9)
Progression way of first-line treatment, n (%)
   Primary lesion 8 (57.1)
   New lesion 6 (42.9)

ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor.

Most patients (11/14, 78.6%) had distant metastasis at baseline of dabrafenib plus trametinib therapy, with the most common sites being contralateral lung (35.7%), bone (21.4%) and brain (21.4%). In the initial genetic analysis, all patients were found to harbor EGFR-sensitive mutations, with 71.4% exhibiting deletions in exon 19 and 21.4% showing the L858R mutation in exon 21. Additionally, one patient displayed both the exon 19 deletion and the exon 21 L858R mutation concurrently. As for first-line therapy, 42.9% of patients received first-generation EGFR-TKIs, 14.2% received second-generation, and 42.9% received third-generation. Eight patients (57.1%) took the primary lesion progression as the progression way of the first line treatment, while the rest of the patients developed the new lesions.

Treatment history

The key findings of 14 patients are summarized in Table 2. Eight patients used first or second-generation EGFR-TKIs as first-line, then switched to osimertinib; six patients chose osimertinib directly as first-line. In all these cases, following treatment with osimertinib, NGS of blood or tissue samples was conducted to detect resistance mechanisms.

Table 2

Targeted therapy regimen and duration of 14 patients on the basis of NGS test

Patient Age, years Sex Histology Stage Surgical history EGFR mutation EGFR-TKI treatment history/time (months) Post-progression NGS BRAF-TKI treatment history/time (months) Therapy response to BRAF-TKI
1 76 M Adeno III Yes EGFR 19del Icotinib/17; osimertinib/10 EGFR 19del; BRAF V600E Dabrafenib + trametinib/15.9 PR
2 61 F Adeno IV No EGFR 19del EFGR L858R Icotinib/29.1; furmonertinib/10.8 EGFR 19del; EGFR L858R; EGFR T790M; BRAF V600E Dabrafenib + trametinib/2.6 SD
3 39 F Adeno IV No EGFR 19del Gefitinib/11.9; osimertinib/16.1 EGFR 19del; EGFR T790M; BRAF V600E Osimertinib + dabrafenib + trametinib/1.2 PD
4 58 M Adeno III Yes EGFR 19del Gefitinib/24; osimertinib/33.8 EGFR 19del; EGFR T790M; BRAF V600E Osimertinib + dabrafenib + trametinib/14.1 PR
5 61 F Adeno IV No EGFR 19del Afatinib/34; osimertinib/7.7 EGFR 19del; BRAF V600E Osimertinib + dabrafenib + trametinib/17.5 PR
6 61 F Adeno III Yes EGFR 19del Icotinib/23.2; furmonertinib/22.8 EGFR 19del; BRAF V600E Furmonertinib + dabrafenib + trametinib/3.3 SD
7 27 F Adeno IV No EGFR 19del Afatinib/12.5; osimertinib/14.1 EGFR 19del; BRAF V600E; TP53 mutation; MET amplification Almonertinib + dabrafenib + trametinib/1.1 SD
8 54 M Adeno II Yes EGFR 19del Gefitinib/31; osimertinib/24.3 EGFR 19del; BRAF V600E; TP53 mutation Osimertinib + dabrafenib + trametinib/2 PD
9 86 M Adeno IV No EGFR 19del Osimertinib/23.5 EGFR 19del; BRAF V600E; TP53 mutation Osimertinib + dabrafenib + trametinib/2.5 SD
10 69 F Adeno IV No EGFR L858R Osimertinib/0.93 EGFR L858R; BRAF V600E Osimertinib + dabrafenib + trametinib/1.1 PD
11 56 F Adeno III Yes EGFR L858R Osimertinib/22.1 EGFR L858R; BRAF V600E Osimertinib + dabrafenib + trametinib/6.7 PR
12 59 F Adeno IV No EGFR 19del Osimertinib/21.2 EGFR 19del; BRAF V600E Osimertinib + dabrafenib + trametinib/8.8 CR
13 52 M Adeno III Yes EGFR L858R Osimertinib/28.5 EGFR L858R; BRAF V600E; EGFR C797S; TP53 mutation Icotinib + dabrafenib + trametinib/7.1 SD
14 70 M Adeno IV No EGFR 19del Osimertinib/17.5 EGFR 19del; BRAF V600E Furmonertinib + dabrafenib + trametinib/7.8 PR

NGS, next-generation sequencing; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; M, male; F, female; PR, partial response; SD, stable disease; PD, progressive disease; CR, complete response.

The second NGS testing results revealed that all cases retained their initial EGFR mutations and additionally acquired new BRAFV600E mutations. It is noteworthy that half of the cases (7/14) showed additional mutations in the second NGS testing, apart from BRAFV600E mutations, including EGFRT790M/C797S mutations, TP53 mutations and MET amplification. As of today, there is no therapeutic strategy to simultaneously inhibit both the EGFR and BRAF/MEK pathways. Therefore, the combination of EGFR-TKI osimertinib, BRAF inhibitor dabrafenib, and MEK inhibitor trametinib represents a rational and feasible treatment approach.

Tumor responses

Tumor responses are shown in Table 3. One patient (7.1%) achieved CR, 4 patients (28.6%) achieved PR and 6 patients (42.9%) had SD, leading to an ORR of 35.7% (95% CI: 14.0–64.4%) and a DCR of 78.6% (95% CI: 52.4–92.4%). Three patients (21.4%) were reported progressive disease (PD) as the best response.

Table 3

Summary of responses

Variables Value
Best overall response, event/total (%)
   CR 1/14 (7.1)
   PR 4/14 (28.6)
   SD 6/14 (42.9)
   PD 3/14 (21.4)
ORR (95% CI), % 35.7 (14.0–64.4)
DCR (95% CI), % 78.6 (52.4–92.4)

CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; ORR, objective response rate; DCR, disease control rate; CI, confidence interval.

A swimmer plot (Figure 2) was generated to facilitate comparison of the duration and therapeutic response across each line of treatment targeting secondary acquired driver alterations in EGFR-mutated NSCLC patients. Time 0 is defined as the initiation of first-line targeted therapy following the identification of the EGFR mutation. The yellow bars indicate the treatment duration with first- or second-generation EGFR-TKIs, the purple bars represent the treatment duration with third-generation TKIs, and the blue bars denote the treatment duration with the addition of BRAF inhibitor dabrafenib and MEK inhibitor trametinib following the second NGS test.

Figure 2 Swimmer plot of the duration and therapeutic response for patients in each line treatment. EGFR-TKI, epidermal growth factor receptor-tyrosine kinase inhibitor; D+T, dabrafenib plus trametinib; CR, complete response; PD, progressive disease; PR, partial response; SD, stable disease.

Clinical outcomes

At the time of the data cutoff (August 1, 2024), the estimated median PFS was 6.7 months (95% CI: 2.5–NE) and 11 (11/14, 78.6%) patients developed disease progression (Figure 3A).

Figure 3 The clinical response of triple regimen, EGFR-TKI plus dabrafenib and trametinib. (A) Kaplan-Meier curve PFS of triple regimen in all 14 patients with newly acquired BRAFV600E mutation after EGFR-TKI resistance. (B) Kaplan-Meier curve PFS of triple regimen treatment in patients receiving different EGFR-TKIs options. (C) Kaplan-Meier curve PFS of triple regimen treatment in patients with different gene mutation backgrounds. (D-F) Kaplan-Meier curve PFS of triple regimen treatment in patients with different surgical history, smoking history and the mode of progression after first-line EGFR-TKI treatment. PFS, progression-free survival; CI, confidence interval; NE, not evaluated; EGFR-TKI, epidermal growth factor receptor-tyrosine kinase inhibitor; D+T, dabrafenib plus trametinib; HR, hazard ratio.

Death occurred in 6 patients (6/14, 42.9%) in all cases. The median OS was not reached in these patients.

Subgroup analysis and potential influencing factors

To explore whether the sequence of EGFR-TKI administration affects patients’ PFS and OS, patients were divided into two groups: one receiving third-generation TKI after first-/second-generation EGFR-TKIs (8 patients) and the other receiving third-generation TKI as first-line treatment (6 patients). The median PFS was 8.35 months in the former group and 6.9 months in the latter, with no significant difference in PFS between the two groups (HR: 1.107; 95% CI: 0.318–3.854; P=0.85) (Figure 3B).

Next, we stratified patients to investigate factors affecting the efficacy of dabrafenib plus trametinib treatment. The presence of additional mutations (e.g., EGFRT790M/C797S, TP53 mutations and MET amplification), after resistance to osimertinib, potentially influenced treatment efficacy. In seven patients with complex mutation profiles, all experienced disease progression, with a median PFS of 2.5 months. However, in seven patients with only BRAF mutations, four of them progressed with a median PFS of 15.9 months (HR: 3.538; 95% CI: 1.008–12.422; P=0.02) (Figure 3C). PFS was not significantly influenced by surgical history (P=0.90, Figure 3D), smoking history (P=0.79, Figure 3E) or the mode of progression after first-line EGFR-TKI treatment (P=0.94, Figure 3F).

The pie chart (Figure 4) visually demonstrated that the group with a complex background, characterized by multiple mutations, showed a significant disadvantage in response with the triple therapy regimen. This highlights that a complex genetic mutation background and individual heterogeneity contribute to the uncertainty of drug efficacy and the early onset of resistance.

Figure 4 Disadvantages in response and early onset of resistance to the triple therapy regimen for patients with complex genetic mutation backgrounds. PD, progressive disease; SD, stable disease; PFS, progression-free survival; CR, complete response; PR, partial response.

Safety

AEs of any grade occurred in 13 patients (93%) and of grade 3 or worse were noted in 4 patients (29%) (Table 4). No unexpected AEs and grade 5 AEs were observed. Pyrexia (8/14, 57%) was the most commonly reported. Apart from pyrexia, gastrointestinal-related adverse reactions (including nausea, diarrhea, weight loss, and decreased appetite) and skin-related adverse reactions (including dry skin, pruritus, and rash) warrant close attention. One patient experienced a dose interruption due to a grade 3 rash. Two patients discontinued dabrafenib plus trametinib because of grade 4 diarrhea and one patient discontinued because of grade 4 pyrexia. Based on the current data, the combination of the three drugs did not lead to fatal adverse reactions and the safety profile was considered manageable.

Table 4

Overview of treatment-related adverse events

Common adverse events Dabrafenib + trametinib (n=14), n [%]
Total Grade 3–4
Any 13 [93] 4 [29]
Pyrexia 8 [57] 1 [7]
Diarrhea 4 [29] 2 [14]
Rash 4 [29] 1 [7]
Nausea 4 [29] 0
Dry skin 4 [29] 0
Pruritus 3 [21] 0
Weight loss 2 [14] 0
Fatigue 1 [7] 0
Decreased appetite 1 [7] 0
Asthenia 1 [7] 0
Peripheral edema 1 [7] 0
Aspartate amino transferase increased 1 [7] 0

Discussion

This retrospective study represents the largest investigation to date on the use of a triple therapy regimen comprising an EGFR-TKI plus dabrafenib and trametinib in patients with advanced NSCLC who developed BRAFV600E mutations after progressing on osimertinib. The study aimed to explore the effectiveness and safety of this triple therapy approach, as well as potential influencing factors that may affect its efficacy including the sequential use of first-, second-, and third-generation EGFR-TKIs, complex genetic mutation backgrounds, history of radical surgery, smoking history, and the progression pattern of first-line therapy.

Newly concomitant BRAFV600E mutations, as a mechanism of resistance to third-generation EGFR-TKIs, pose a significant challenge in the treatment of advanced NSCLC. Because secondary BRAF drivers identified after progression on EGFR-TKIs are rare, and molecular alterations and associated therapies are highly heterogeneous, no prospective trials have yet reported the efficacy of this approach. Consequently, treatment decisions must currently be guided by case reports and case series. Giustini et al. (15) described in their case series of 8 patients that all of them retained their original EGFR mutations at the time of post-progression NGS, consistent with the cases we have observed in clinical practice, explaining why osimertinib is recommended to combine with the dabrafenib and trametinib regimen.

Given that all patients in our study exhibited BRAFV600E mutations after osimertinib failure, this underscores the importance of utilizing NGS to identify resistance mechanisms following osimertinib failure. However, in clinical practice, it is important to note that the accuracy and reliability of NGS results are influenced by multiple factors including sample source, quality, the sequencing platform and so on (16,17). The case report by Chimbangu et al. (18) highlighted that a patient developed resistance to Gefitinib and no actionable driver mutations were detected in the peripheral blood. Consequently, the treatment was switched to chemotherapy and a subsequent liver biopsy revealed a BRAF mutation. This highlights the importance of high-quality biopsy samples and appropriate timing of testing for NGS results, which directly impact patient treatment decisions.

In previous research conducted by Wei et al. (19), it was demonstrated that the median PFS for 5 patients treated with a combination of EGFR and BRAF inhibitors [vemurafenib (n=4); dabrafenib plus trametinib (n=1)] was 7.8 months, whereas it was only 5.0 months for 17 patients receiving chemotherapy. In our study, the median PFS for 14 patients with acquired BRAFV600E mutations undergoing simultaneous EGFR and BRAF inhibition was 6.7 months. Several studies (20,21) have indicated that for patients with EGFR-TKI resistance, the efficacy of immunotherapy combined with chemotherapy does not surpass that of chemotherapy alone. However, adding anti-angiogenic agents to the immunotherapy-chemotherapy combination, as demonstrated by the ORIENT-31 and ATTLAS trials (22,23), suggests that a four-drug regimen enhance PFS benefits. Nonetheless, the associated tolerability and safety concerns of the four-drug regimen pose new challenges. Therefore, as a precision treatment strategy, the triple regimen of EGFR-TKI plus dabrafenib and trametinib can be considered as an alternative option before resorting to cytotoxic chemotherapy regimens for such patients. An interesting finding we observed is that BRAFV600E mutation was not detected after patients developed resistance to first- or second-generation EGFR-TKIs, but after resistance to osimertinib, so it remains unclear whether patients treated with osimertinib are more prone to developing acquired BRAF mutations compared to those treated with other EGFR-TKIs. Additionally, the use of multiple EGFR-TKIs does not significantly impact the PFS of the triple regimen after progression.

The phenomenon of resistance to EGFR-TKI therapy and its underlying mechanisms have been extensively studied and discussed. Feldt et al. (24) and Michels et al. (25) focused on how MET amplification impacts the efficacy of EGFR inhibitors and explored how concurrent mutations further diminish therapeutic effectiveness. Hernández Borrero and El-Deiry (26) and Stiewe and Haran (27) have reviewed how the p53 pathway functions as a complex cellular stress response network that interacts with the genome to promote tumorigenesis and drug resistance. Based on our study cohort, MET amplification, TP53 mutations, and the T790M mutation are identified as key factors contributing to the increased genetic complexity in patients with EGFR resistance who subsequently develop BRAFV600E mutations. Although the statistical significance has not yet reached, this may be a potential reason why patients with complex genetic mutation backgrounds exhibit poorer efficacy and survival outcomes. This finding could provide valuable insights for future clinical diagnosis and treatment.

Furthermore, the safety profile of the triple regimen is a crucial consideration and a prerequisite for its potential adoption. In addition to pyrexia, which has been identified as the most frequent AE in multiple studies (28,29), attention should also be given to gastrointestinal and dermatological adverse effects. Notably, based on the current research data, the combination of these three drugs does not lead to an additive effect in terms of AEs when compared to their individual use. The incidence of grade ≥3 AEs is comparable to those observed with standard therapies and no unexpected toxicities are identified, indicating an overall manageable safety profile.

There are several limitations in this study, including its retrospective nature, the inconsistency in sources and methodologies for secondary or tertiary NGS samples, and the small sample size due to the rarity of BRAF mutations.


Conclusions

In conclusion, the triple therapy regimen of EGFR-TKI plus dabrafenib and trametinib was found to have substantial and durable clinical benefit, with a manageable safety profile, in patients with newly concomitant BRAFV600E mutations after osimertinib failure. However, the presence of a complex background with multiple genetic mutations may potentially reduce the efficacy of this regimen.


Acknowledgments

Funding: This work was supported by Shanghai Municipal Health Commission (No. 2020CXJQ02), Shanghai Anti-Cancer Association (No. SACA-AX202210), Shanghai Municipal Science and Technology Commission (No. 22Y31920405), Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases (No. 20dz2261100), Shanghai Municipal Key Clinical Specialty (No. shslczdzk02202) and Beijing Life Oasis Public Service Center (No. CPHCF-ZLKY-2023016).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-803/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-803/dss

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-803/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-803/coif). The authors have no 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. This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013), and the protocol was reviewed and approved by the institutional review board of Ruijin Hospital (No. 2024-164), the Ethics Committee of Fudan University Shanghai Cancer Center (No. 1612167-18) and the Ethics Committee of Shanghai Pulmonary Hospital (No. L22-407). Individual consent for this retrospective analysis was waived.

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

  1. Leiter A, Veluswamy RR, Wisnivesky JP. The global burden of lung cancer: current status and future trends. Nat Rev Clin Oncol 2023;20:624-39. [Crossref] [PubMed]
  2. Hendriks LE, Kerr KM, Menis J, et al. Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023;34:339-57. [Crossref] [PubMed]
  3. Riely GJ, Wood DE, Ettinger DS, et al. Non-Small Cell Lung Cancer, Version 4.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2024;22:249-74. [Crossref] [PubMed]
  4. Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol 2011;29:2866-74. [Crossref] [PubMed]
  5. Yang JC, Sequist LV, Geater SL, et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol 2015;16:830-8. [Crossref] [PubMed]
  6. 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]
  7. Jia Y, Yun CH, Park E, et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature 2016;534:129-32. [Crossref] [PubMed]
  8. Ferro A, Marinato GM, Mulargiu C, et al. The study of primary and acquired resistance to first-line osimertinib to improve the outcome of EGFR-mutated advanced Non-small cell lung cancer patients: the challenge is open for new therapeutic strategies. Crit Rev Oncol Hematol 2024;196:104295. [Crossref] [PubMed]
  9. Saalfeld FC, Möller J, Christopoulos P, et al. Small cell transformation in EGFR-mutated non-small cell lung cancer: DLL3 expression and efficacy of immune checkpoint inhibitors or tyrosine kinase inhibitors combined with chemotherapy. Eur J Cancer 2024;213:115065. [Crossref] [PubMed]
  10. Sui JSY, Ladanyi M, Rudin CM, et al. Clinicopathologic and mutational landscape of BRAFV600E-mutant non–small cell lung carcinoma. J Clin Oncol 2022;40:9084.
  11. Chen J, Facchinetti F, Braye F, et al. Single-cell DNA-seq depicts clonal evolution of multiple driver alterations in osimertinib-resistant patients. Ann Oncol 2022;33:434-44. [Crossref] [PubMed]
  12. Aboubakar Nana F, Ocak S. Targeting BRAF Activation as Acquired Resistance Mechanism to EGFR Tyrosine Kinase Inhibitors in EGFR-Mutant Non-Small-Cell Lung Cancer. Pharmaceutics 2021;13:1478. [Crossref] [PubMed]
  13. La Monica S, Minari R, Cretella D, et al. Acquired BRAF G469A Mutation as a Resistance Mechanism to First-Line Osimertinib Treatment in NSCLC Cell Lines Harboring an EGFR Exon 19 Deletion. Target Oncol 2019;14:619-26. [Crossref] [PubMed]
  14. Odogwu L, Mathieu L, Blumenthal G, et al. FDA Approval Summary: Dabrafenib and Trametinib for the Treatment of Metastatic Non-Small Cell Lung Cancers Harboring BRAF V600E Mutations. Oncologist 2018;23:740-5. [Crossref] [PubMed]
  15. Giustini NP, Patel SP, Myall NJ, et al. Resistance to EGFR Tyrosine Kinase Inhibitor Therapy in Non-Small-Cell Lung Cancer via Newly Acquired Targetable Oncogenic Driver Alterations With an Emphasis on BRAF: Case Series and Literature Review of Treatment. JCO Precis Oncol 2022;6:e2100551. [Crossref] [PubMed]
  16. Duan H, Li X, Mei A, et al. The diagnostic value of metagenomic next-generation sequencing in infectious diseases. BMC Infect Dis 2021;21:62. [Crossref] [PubMed]
  17. De Las Casas LE, Hicks DG. Pathologists at the Leading Edge of Optimizing the Tumor Tissue Journey for Diagnostic Accuracy and Molecular Testing. Am J Clin Pathol 2021;155:781-92. [Crossref] [PubMed]
  18. Chimbangu CT, Ya Z, Xi L, et al. Promising response of dabrafenib, trametinib, and osimertinib combination therapy for concomitant BRAF and EGFR-TKI resistance mutations. Anticancer Drugs 2024;35:109-15. [Crossref] [PubMed]
  19. Wei XW, Deng JY, Xu CR, et al. Characteristics of and Treatment Strategies for Advanced EGFR-Mutant NSCLC With Concomitant BRAF Variations. JTO Clin Res Rep 2022;3:100348. [Crossref] [PubMed]
  20. 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] [PubMed]
  21. 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]
  22. Lu S, Wu L, Jian H, et al. VP9-2021: ORIENT-31: Phase III study of sintilimab with or without IBI305 plus chemotherapy in patients with EGFR mutated nonsquamous NSCLC who progressed after EGFR-TKI therapy. Ann Oncol 2022;33:112-3.
  23. Park S, Kim TM, Han JY, et al. Phase III, Randomized Study of Atezolizumab Plus Bevacizumab and Chemotherapy in Patients With EGFR- or ALK-Mutated Non-Small-Cell Lung Cancer (ATTLAS, KCSG-LU19-04). J Clin Oncol 2024;42:1241-51. [Crossref] [PubMed]
  24. Feldt SL, Bestvina CM. The Role of MET in Resistance to EGFR Inhibition in NSCLC: A Review of Mechanisms and Treatment Implications. Cancers (Basel) 2023;15:2998. [Crossref] [PubMed]
  25. Michels S, Heydt C, van Veggel B, et al. Genomic Profiling Identifies Outcome-Relevant Mechanisms of Innate and Acquired Resistance to Third-Generation Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy in Lung Cancer. JCO Precis Oncol 2019;3:PO.18.00210.
  26. Hernández Borrero LJ, El-Deiry WS. Tumor suppressor p53: Biology, signaling pathways, and therapeutic targeting. Biochim Biophys Acta Rev Cancer 2021;1876:188556. [Crossref] [PubMed]
  27. Stiewe T, Haran TE. How mutations shape p53 interactions with the genome to promote tumorigenesis and drug resistance. Drug Resist Updat 2018;38:27-43. [Crossref] [PubMed]
  28. Planchard D, Besse B, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol 2016;17:984-93. [Crossref] [PubMed]
  29. Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017;18:1307-16. [Crossref] [PubMed]
Cite this article as: Yang W, Feng X, Ni J, Zhang X, Yu H, Wu X, Wang H, Zhao X, Hu Z, Yu B, Zhang Y, Lin Y, Xiang Y, Wang J. EGFR inhibitors plus dabrafenib and trametinib in patients with EGFR-mutant lung cancer and resistance mediated by BRAFV600E mutation: a multi-center real-world experience in China. Transl Lung Cancer Res 2024;13(12):3500-3512. doi: 10.21037/tlcr-24-803

Download Citation