Skip to main content
Erschienen in: Targeted Oncology 3/2023

Open Access 05.04.2023 | Original Research Article

Genomic Landscape of Primary Resistance to Osimertinib Among Hispanic Patients with EGFR-Mutant Non-Small Cell Lung Cancer (NSCLC): Results of an Observational Longitudinal Cohort Study

verfasst von: Diego F. Chamorro, Andrés F. Cardona, July Rodríguez, Alejandro Ruiz-Patiño, Oscar Arrieta, Darwin A. Moreno-Pérez, Leonardo Rojas, Zyanya Lucia Zatarain-Barrón, Dora V. Ardila, Lucia Viola, Gonzalo Recondo, Juan B. Blaquier, Claudio Martín, Luis Raez, Suraj Samtani, Camila Ordóñez-Reyes, Juan Esteban Garcia-Robledo, Luis Corrales, Carolina Sotelo, Luisa Ricaurte, Mauricio Cuello, Sergio Mejía, Elvira Jaller, Carlos Vargas, Hernán Carranza, Jorge Otero, Pilar Archila, Maritza Bermudez, Tatiana Gamez, Alessandro Russo, Umberto Malapelle, Diego de Miguel Perez, Vladmir C. Cordeiro de Lima, Helano Freitas, Erick Saldahna, Christian Rolfo, Rafael Rosell, CLICaP

Erschienen in: Targeted Oncology | Ausgabe 3/2023

Abstract

Background

Epidermal growth factor receptor (EGFR) mutations (EGFRm) represent one of the most common genomic alterations identified among patients with non-small cell lung cancer (NSCLC). Several targeted agents for patients with EGFRm have been proven safe and effective, including the third-generation tyrosine kinase inhibitor (TKI) osimertinib. Nonetheless, some patients will present with or develop EGFR-TKI resistance mechanisms.

Objective

We characterized the genomic landscape of primary resistance to osimertinib among Hispanic patients with EGFR-mutant NSCLC.

Methods

An observational longitudinal cohort study was conducted with two groups of patients, those with intrinsic resistance (cohort A) and those with long-term survival (cohort B). All patients were treated and followed between January 2018 and May 2022. All patients were assessed for Programmed Cell Death Ligand 1 (PD-L1) expression and Bcl-2-like protein 11 (BIM)/AXL mRNA expression before starting TKI. After 8 weeks of treatment, a liquid biopsy was performed to determine the presence of circulating free DNA (cfDNA), and next-generation sequencing (NGS) was used to identify mutations at the time of progression. In both cohorts, overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) were evaluated.

Results

We found a homogeneous distribution of EGFR-sensitizing mutations in both cohorts. For cohort A, exon 21 mutations were more common than exon 19 deletions (ex19dels) for cohort B (P = 0.0001). The reported ORR for osimertinib was 6.3% and 100% for cohorts A and B, respectively (P = 0.0001). PFS was significantly higher in cohort B (27.4 months vs. 3.1 months; P = 0.0001) and ex19del patients versus L858R (24.5 months, 95% confidence interval [CI] 18.2–NR), vs. 7.6 months, 95% CI 4.8–21.1; P = 0.001). OS was considerably lower for cohort A (20.1 months vs. 36.0 months; P = 0.0001) and was better for patients with ex19del, no brain metastasis, and low tumor mutation burden. At the time of progression, more mutations were found in cohort A, identifying off-target alterations more frequently, including TP53, RAS, and RB1.

Conclusion

EGFR-independent alterations are common among patients with primary resistance to osimertinib and significantly impact PFS and OS. Our results suggest that among Hispanic patients, other variables associated with intrinsic resistance include the number of commutations, high levels AXL mRNA, and low levels of BIM mRNA, T790M de novo, EGFR p.L858R presence, and a high tumoral mutational burden.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s11523-023-00955-9.
Diego F. Chamorro, Andrés F. Cardona, July Rodríguez, Alejandro Ruiz-Patiño, Oscar Arrieta contributed equally to the study.
CLICaP: Latin American Consortium for the Investigation of Lung Cancer.
Key Points
The prognosis of patients with EGFR mutations and primary resistance to osimertinib is utterly opposed to those with a high sensitivity to osimertinib.
Patients with EGFR mutations with complex genotypes, including alterations in TP53 and RB1, and a high tumor mutation burden, require a different therapeutic approach to osimertinib as the first-line intervention.
Early assessment of EGFR circulating free DNA (cfDNA) allows prediction of the disease course in patients with primary resistance to osimertinib. This information could allow the use of risk-adaptive therapeutic models.
AXL mRNA expression levels are considerably higher among patients with EGFR mutations and primary resistance to osimertinib. In contrast, Bcl-2 interacting mediator of cell death (BIM) mRNA expression levels were significantly lower in cohort A.

1 Introduction

Alterations in the epidermal growth factor receptor (EGFR) gene are among the most common oncogenic driver mutations in the pathogenesis of non-small cell lung cancer (NSCLC). Specific mutations in the EGFR gene are associated with increased sensitivity to EGFR tyrosine kinase inhibitors (EGFR-TKI), which offer the best possible results for treatment response in patients with an NSCLC diagnosis and EGFR mutations (EGFRm) [1, 2]. EGFRm presence has been reported in 10–50% of NSCLC cases, almost all in lung adenocarcinomas (LUADs). In addition, significant ethnic variations for EGFRm prevalence have been described, such as 8–15% in LUADs diagnosed in Caucasian patients versus 30–50% in East Asian populations [3, 4]. Among Hispanic populations in Latin America, the frequency of EGFRm in LUADs ranges from 14% in Argentina to 24–35% in Colombia and as high as 50% among Peruvian populations [57]. Evaluation of ancestry–mutation association showed that Native American ancestry is positively correlated with EGFRm [8, 9].
Among the EGFR-TKI-sensitizing mutations, exon 19 deletions (ex19dels) in the p.E746-A750 region and the point-substitution p.L858R (L858R) in exon 21 are the two most commonly described [10, 11]. These alterations represent ≈90% of EGFR-TKI-sensitizing mutations [12]. ex19dels and L858R are early clonal events that lead to tumor development in LUADs. They result in constitutive ligand-independent EGFR-tyrosine kinase activity and an increased affinity for first-generation (i.e., gefitinib, erlotinib, and icotinib), second-generation (i.e., afatinib and dacomitinib), and third-generation (i.e., osimertinib and aumolertinib) EGFR-TKI [13]. Nowadays, the National Comprehensive Cancer Network recommends osimertinib as the preferred EGFR-TKI option as first-line treatment for NSCLC because it inhibits both EGFR-TKI-sensitizing and the EGFR resistance mutation p.Thr790Met (T790M), and it has shown efficacy in patients with central nervous system (CNS) metastases [14].
The FLAURA trial (NCT02296125) recruited over 550 patients with ex19dels or L858R EGFR-mutated advanced NSCLC, from December 2014 to March 2016 [15]. In this study, the individuals were stratified according to EGFRm status and ethnicity (Asian/non-Asian), then half of the patients were treated with oral osimertinib and the other half were treated with other TKI [1618]. After follow-up, the study demonstrated a better overall survival (OS) and a favorable OS rate in the group treated with osimertinib. Although the FLAURA trial and other studies have consistently shown the superior efficacy of osimertinib in comparison with first- and second-generation EGFR-TKI, some patients develop resistance mechanisms to third-generation TKI. These resistance mechanisms affect a patient's prognosis under TKI treatment. Most patients with advanced EGFRm NSCLC will achieve an objective response after treatment with TKI, but will eventually present disease progression. The extent and duration of responses are variable. However, approximately 10–15% of patients do not respond to treatment or have control of the disease for a short time, usually less than 3 months [19, 20].
Current evidence suggests two types of resistance mechanisms: EGFR dependent and EGFR independent. Some EGFR-dependent resistance mechanisms have been described for first-line and second-line treatment, including acquired EGFRm (C797S, L718/G719, G796/C797, L792, L798, and EGFR amplification). These profiles pose a significant challenge in selecting the most effective treatment based on resistance patterns after disease progression. Regarding the EGFR-independent mechanisms of acquired resistance, the activation of bypass pathways via amplifications (i.e., epithelial growth factor receptor or ERBB2/HER2, MET, fibroblast growth factor receptor 1 or FGFR1, and KRAS), fusions of alternative receptor tyrosine kinase (RTK) genes (i.e., RET, anaplastic lymphoma kinase [ALK], fibroblast growth factor receptor 3 [FGFR3], and neurotrophic receptor tyrosine kinase 1 [NTRK1]), as well as activating mutations and fusions of members of the downstream RAS-RAF-MEK-MAPK and PI3K/AKT/PTEN/mTOR pathways have been proposed as possible explanations [21]. Additional mechanisms of acquired resistance shared by TKI of all three generations include the phenotypic transformation to small-cell lung cancer (SCLC), epithelial-mesenchymal transition (EMT), the conversion to squamous cell carcinoma (SCC), activation of the Wnt/β-catenin pathway, hormonal signaling, and cell cycle were observed in some EGFR mutant cases compared to wild-type EGFR cases [2225].
It has been proposed that acquired resistance could emerge from the expansion of original pre-treatment resistant clones and new resistance mechanisms developed as an adaptative response to TKI exposure. On the other hand, intrinsic resistance mechanisms are still a matter of debate and secondary activating pathways remain poorly understood [19, 20, 26]. Herein, we characterize the clinical, molecular, and genomic features associated with an intrinsic resistance pattern to osimertinib therapy in treatment-naïve patients with EGFRm NSCLC [24, 25].

2 Methods

2.1 Study Design and Patients

An observational longitudinal cohort study was conducted by the Latin American Consortium for Investigation of Lung Cancer (CLICaP) using the data deposited in a centralized database (CLICaP Real World Data/Evidence database). Patients included in the EGFRm NSCLC database were assessed from January 2018 to May 2022. All patients were residents of Bogota, DC, Colombia, and demographic variables such as age, sex, and smoking history were collected from the clinical record. Eligible patients were those with EGFRm analyzed by molecular diagnosis (N = 578) (Fig. 1). Among these patients, we defined cohort A as cases with intrinsic resistance to first-line osimertinib with measurable disease progression within the first 3 months of treatment (N = 16). In contrast, cohort B were patients with long-term responses to osimertinib treatment or progression-free survival (PFS) with osimertinib > 18 months. Finally, this control group was selected from a cohort of long survivors with genomic evaluation by next-generation sequencing (NGS) and complete clinical follow-up. Cases with similar clinical characteristics were sought without being able to match them, due to the unusual nature of patients with primary resistance.
At diagnosis, tumor tissue was acquired from all included patients to undergo molecular characterization, including PD-L1 expression by immunohistochemistry (IHC) and a standardized NGS test. A liquid biopsy was also obtained before starting osimertinib to evaluate Bcl-2 interacting mediator of cell death (BIM) polymorphisms. In the baseline tumor tissue, BIM and AXL mRNA expression levels were quantified using reverse transcription-polymerase chain reaction (RT-PCR). Cohort A and B patients received first-line osimertinib (80 mg orally once daily) until disease progression or unacceptable toxicity was documented. Patients were followed for 48 months and assessed for progression. After 8 weeks of treatment with osimertinib, a new liquid biopsy was performed to estimate the EGFRm status by droplet digital PCR (ddPCR). The NGS test in liquid biopsy was repeated when progression was found, and tissue collection was attempted when there was suspicion of histological transformation (SCLC). The study design and patient inclusion are summarized in Fig. 1.

2.2 Follow-Up and Clinical Outcomes

Patients included in the study were assessed clinically every month. Radiological follow-ups were completed every 9–12 weeks. Clinical, demographic, and molecular variables (including all mutations and tumor mutational burden [TMB]) were stored in a centralized and deidentified database at the CLICaP/Foundation for Clinical and Applied Cancer Research—FICMAC (Bogotá, Colombia). All included patients provided signed informed consent. In addition, an Institutional Review Board and Privacy Board waiver was obtained to facilitate retrospective clinical-pathological and molecular data (Lung Cancer-FICMAC/CLICaP Platform—Registration No. 2012/014, Kayre, Bogotá, Colombia, and Research Ethics Committee of Universidad El Bosque—Registration No. PCI-2018-10171).
Overall response rate (ORR) was defined as the sum of complete responses (CRs) and partial responses (PRs), as assessed by an independent, blinded radiologist according to the Response Evaluation Criteria in Solid Tumors (RECIST) V1.1. For PFS, time to event was calculated from the date of treatment start until radiographic disease progression as assessed by a blinded radiologist according to RECIST V1.1 or death by any cause. Observations for patients who did not experience progression were censored at the patient-specific last follow-up. OS was estimated from diagnosis until death by any cause or loss to follow-up.

2.3 PD-L1 Testing

PD-L1 expression was determined by IHC using the Dako 22C3 pharmDx kit, considering more than 100 tumor cells in the slide section for accurate PD-L1 readings. PD-L1 testing was completed on biopsies taken at diagnosis. For survival analyses, patients were stratified according to PD-L1 status (i.e., ≥ 50%, 1–49%, and < 1% subgroups). Patients with unknown PD-L1-expression status were also included in this study.

2.4 cfDNA Detection (Week 8) by Droplet Digital PCR Analysis

The first blood sample was collected at the time of diagnosis before the start of treatment. Circulating free DNA (cfDNA) was quantified using the QX200 Droplet Digital PCR System (Bio-Rad Laboratories, Hercules, CA). For the EGFR multiplex assays, a final PCR mix volume of 20 μL was manually loaded into wells of a DG8 cartridge (Bio-Rad Laboratories) with 70 μL of Droplet Generation Oil for probes (Bio-Rad Laboratories). After droplet generation by the QX200 Droplet Generator (Bio-Rad Laboratories), 40 μL of the sample was transferred into a 96-well PCR plate and amplified with a C1000 Touch Thermal Cycler (Bio-Rad Laboratories), using the following thermal cycling conditions: 95 °C for 10 min, 40 cycles at 94 °C for 30 s, 55 °C for 1 min (2 °C/s), and 98 °C for 10 min, and a final cooling step to 12 °C (1 °C/s). The droplets were analyzed using the QX200 Droplet Reader (Bio-Rad Laboratories), and data were analyzed using QuantaSoft software version 1.7.4.0917 and QuantaSoft Analysis Pro software version 1.0.596 (Bio-Rad Laboratories). Thresholds were placed manually, and the fractions of positive and negative droplets were used to calculate the concentration and fractional abundance of target DNA sequences with their 95% Poisson-based confidence intervals (CI). EGFRm primers and probes were based on previous studies [27, 28].

2.5 AXL and BIM mRNA Quantification by RT-PCR Analyses

Samples were lysed in a buffer containing EDTA, SDS, and proteinase K. Then RNA was extracted with phenol-chloroform-isoamyl alcohol, followed by precipitation with isopropanol in the presence of glycogen and sodium acetate. RNA was homogenized in water and treated with DNAse I to avoid DNA contamination. Complementary DNA (cDNA) was synthesized using M-MLV (Moloney Murine Leukemia Virus Reverse Transcriptase) retro-transcriptase enzyme. cDNA was added to Taqman Universal Master Mix in a 12.5-μL reaction with specific primers and probes for each gene. The primer and probe sets were designed using the LightCycler® Probe Design Software 2.0 (Roche Diagnostics Corporation, Indianapolis, Indiana) according to their Ref Seq (National Center for Biotechnology Information). Quantification of gene expression was performed using the LightCycler® 480 Real-Time PCR System (Roche Diagnostics Corporation, Indianapolis, Indiana) and calculated according to the comparative Ct method. The final results were determined as follows: 2-(ΔCt sample-ΔCt calibrator), where ΔCt values of the calibrator and sample are determined by subtracting the Ct value of the target gene from the value of the endogenous gene (β-actin). Commercial RNA controls were used as calibrators (Stratagene, La Jolla, CA, USA). In quantitative experiments, a sample was considered non-viable when the standard deviation (SD) of the Ct values was ≥ 0.30 in two independent analyses. To dichotomize the mRNA expression levels, AXL 1.42 (high AXL [AXL-H] ≥ 1.42) and BIM 0.98 (high BIM [BIM-H] ≥ 0.98) were used as cut-off points; the cut-off points are based on the values described in the existing evidence [29, 30].

2.6 Next-Generation Sequencing

cDNA synthesis prior to library preparation for the RNA panel was carried out using SuperScript™ VILO™ cDNA Synthesis Kit (Thermo Fisher Scientific, 2018, 11754050). Library preparation was carried out using the Oncomine™ Comprehensive Assay Plus (Thermo Fisher Scientific, A49667) and RNA Oncomine™ Fusion's assay (Thermo Fisher Scientific) kits following the instructions of the manufacturer and using a total of 10 ng input DNA and/or RNA per sample (minimum 0.83 ng/μL sample DNA concentration). A maximum of seven DNA samples were prepared per run (six samples if both DNA and RNA analyses were required) on an Ion 540™ Chip (Thermo Fisher Scientific, A27765) using the Ion S5™ sequencing system (see the electronic supplementary material, Supplementary Data 1 and 2). The DNA panel can identify hotspot mutations and copy number variations (CNVs), full coding DNA sequences (CDSs), and RNA fusions (Supplementary Table 1 shows the genes and alterations included in the analysis). Template preparation was performed on the Ion Chef System (Thermo Fisher Scientific) using the Ion PGM Hi-Q Chef Kit and/or the Ion One Touch™ 2 System using the Ion PGM Template OT2 200 Kit. Sequencing was performed using the Ion PGM Hi-Q Sequencing Kit on the Ion Torrent Personal Genome Machine. TMB was measured by running the Oncomine Tumor Mutation Load Assay (Thermo Fisher Scientific, A37909), which enables accurate measurements of somatic mutations without needing a matched normal sample. This assay covers a large genomic footprint of 1.7 Mb, encompassing 1.2 Mb of exonic sequence, to enable accurate mutation counts for samples with a range of TMBs. For the analysis, TMB–high (TMB-H) was defined as ≥ 10 mutations/megabase [mut/Mb]) using the same cut-off as in the KEYNOTE-158 study (NCT02628067) [31].

2.7 Next-Generation Sequencing Data Analysis

Ion Torrent Suite™ Browser version 5.0 and Ion Reporter™ version 5.0 were used to analyze the NGS data. The Torrent Suite™ Browser was used to perform initial quality control, including chip loading density, median read length, and the number of mapped reads. The Coverage Analysis plugin was applied to all data to assess amplicon coverage for regions of interest. Variants were identified by the Ion Reporter filter chain 5% Oncomine™ Variants (5.0). A cut-off of 500X coverage was applied to all analyses. All identified variants were checked for correct nomenclature using Alamut Visual v.2.7.1 (Interactive Biosoftware). Any discrepancies in variant identification between Ion Reporter and Alamut were validated manually using the Integrative Genomics Viewer and NextGENe® v2.4.2 (SoftGenetics®) [32, 33].

2.8 Statistical Analysis

All analyses were conducted on IBM SPSS Statistics software version 25.0 (SPSS Inc. Chicago, IL, USA). Descriptive analyses were used to provide an overview of the study population characteristics. Categorical variables were assessed via the Chi-square test or, whenever appropriate, Fisher’s Exact test. OS and PFS were reported as Kaplan–Meier survival curves. Univariable regression analysis models were generated to assess potential confounders. Mann–Whitney U test with a two-sided P value was established to determine statistically significant outcomes. There were no adjustments made for multiple comparisons, and in all cases, the significance level was 0.05.

3 Results

3.1 Baseline Patient Characteristics

The clinical information derived from 32 Hispanic patients with NSCLC and EGFRm is summarized in Table 1. Demographic characteristics indicate that all patients were diagnosed at stage IV, and most patients had an adenocarcinoma histology type (96.9%), mainly solid or lepidic patterns (56.3%). There is a higher proportion of female patients in both cohorts (60%), and at least 65% had a negative smoking history. Most patients had pleuropulmonary compromise (87.5%) and good functional status (Eastern Cooperative Oncology Group, ECOG performance status 0–1, 90.7%). Samples were obtained from the lung or pleural space in 90.6%. Most patients had a liquid biopsy at progression (96.9%), and in two cases, tumor tissue was obtained by repeated biopsy. Both cases confirm the suspicion of differentiation to SCLC. Overall, the distribution of EGFR-sensitizing mutations was comparable in the population, including ex19dels and L858R mutations. However, patients in cohort A had significantly more frequent alterations in exon 21 than in cohort B in exon 19 (P = 0.0001). Seven patients had secondary EGFRm (21.9%), which occurred more frequently in cohort B (P = 0.32). A baseline T790M mutation was identified in five cases, four in cohort A (P = 0.16), with a mean allele frequency of 39 (SD ± 20). Thirteen patients (87.5%) in cohort A had more than three co-mutations at the time of diagnosis, a significantly higher finding than for cohort B (P = 0.0001). Almost all baseline TP53 commutations (82.4%) and those involving the RAF pathway were identified among patients in cohort A (P = 0.001 and P = 0.009). Besides EGFR and TP53 genes, the main commutations identified at baseline included RB1, AT-rich interactive domain-containing protein 1A [ARID1A], and BRAF. Fig. 2 shows the primary baseline genomic alterations identified in both cohorts, and Supplementary Table 2 details the variants detected at diagnosis (see the electronic supplementary material).
Table 1
Baseline characteristics of patients included in the study
Variable
Overall population (N = 32)
Cohort A (N = 16)
Cohort B (N = 16)
Age at diagnosis
 Mean ± SD
58 ± 10.7
61 ± 12.1
55 ± 8.4
 Median (range)
60 (32–79)
64 (32–79)
51 (44–69)
Sex
 Male
13 (40.6)
8 (50.0)
5 (31.3)
 Female
19 (59.4)
8 (50.0)
11 (68.8)
Smoking history
 Never smoker
20 (62.5)
8 (50.0)
12 (75.0)
 Former smoker
11 (34.4)
7 (43.8)
4 (25.0)
 Current smoker
1 (3.1)
1 (6.3)
Histology at diagnosis
 Adenocarcinoma
31 (96.9)
15 (93.8)
16 (100.0)
 NOS
1 (3.1)
1 (6.3)
Histological subtype
 Papillary
5 (15.6)
1 (6.3)
4 (25.0)
 Micropapillary
4 (12.5)
3 (18.8)
1 (6.3)
 Solid
12 (37.5)
9 (56.3)
3 (18.8)
 Lepidic
6 (18.8)
1 (6.3)
6 (37.5)
 Acinar
4 (12.5)
1 (6.3)
2 (12.5)
 ND
1 (3.1)
1 (6.3)
Baseline tissue origin
 Liver
1 (3.1)
1 (6.3)
 Lung
24 (75.0)
12 (75.0)
12 (75.0)
 Pleura
5 (15.6)
3 (18.8)
2 (12.5)
 Lymph node
1 (3.1)
1 (6.3)
 Small bowel/peritoneum
1 (3.1)
1 (6.3)
ECOG
 0
10 (31.3)
1 (6.3)
9 (56.3)
 1
19 (59.4)
13 (81.3)
6 (37.5)
 2
3 (9.4)
2 (12.5)
1 (6.3)
Number of metastatic sites
 < 2
24 (75%)
9 (56.3)
15 (93.8)
 > 3
8 (25%)
7 (43.8)
1 (6.3)
Brain metastases (baseline)
 Yes
10 (31.3)
9 (56.3)
1 (6.3)
 No
22 (68.8)
7 (43.8)
15 (93.8)
Lung/pleural metastases
 Yes
28 (87.5)
14 (87.5)
14 (87.5)
 No
4 (12.5)
2 (12.5)
2 (12.5)
Liver metastases
 Yes
10 (31.3)
10 (62.5)
 No
22 (68.8)
6 (37.5)
16 (100.0)
Adrenal metastases
 Yes
3 (9.4)
2 (12.5)
1 (6.3)
 No
29 (90.6)
14 (87.5)
15 (93.8)
Bone metastases
 Yes
4 (12.5)
1 (6.3)
3 (18.8)
 No
28 (87.5)
15 (93.8)
13 (81.3)
EGFR mutation
 Exon 19 deletion
15 (46.9)
2 (12.5)
13 (81.3)
 L858R
16 (50.0)
13 (81.3)
3 (18.8)
 G719S
1 (3.1)
1 (6.3)
Secondary EGFR mutations
 Exon 18 p.V869M
1 (3.1)
1 (6.3)
 Exon 21 p.N826S
1 (3.1)
1 (6.3)
 Exon 21 p.K846R
1 (3.1)
1 (6.3)
 Exon 21 p.L861Q
1 (3.1)
1 (6.3)
 Exon 21 p.G863D
1 (3.1)
1 (6.3)
 Exon 19 p.D761Y
1 (3.1)
1 (6.3)
 Exon 19 p.L747S
1 (3.1)
1 (6.3)
T790M mutation at baseline
 Yes
5 (15.6)
4 (25.0)
1 (6.3)
 No
27 (84.4)
12 (75.0)
15 (93.8)
Number of commutations at baseline
 < 2
18 (56.3)
2 (12.5)
16 (100.0)
 ˃ 3
14 (43.8)
14 (87.5)
Baseline commutation in P53
 Yes
17 (53.1)
14 (87.5)
3 (18.8)
 No
15 (46.9)
2 (12.5)
13 (81.3)
Baseline commutations in RAF pathway
 Yes
6 (18.8)
6 (37.5)
 No
26 (81.3)
10 (62.5)
16 (100.0)
PD-L1 expression (at baseline)
 <1%
9 (28.1)
3 (18.8)
6 (37.5)
 1–49%
21 (65.0)
11 (68.8)
10 (62.5)
 ˃ 50%
2 (6.3)
2 (12.5)
TMB at baseline
 Mean TMB
4 (±3.0)
1.3 (±0.5)
1.0 (±1.0)
 TMB-H
5 (15.6)
5 (31.3)
 TMB-L
22 (68.8)
11 (68.8)
11 (68.8)
 ND
5 (15.6)
 
5 (31.3)
Post-osimertinib specimens
 Tumor tissue
2 (6.2)
2 (12.6)
 cfDNA
31 (96.9)
16 (100.0)
15 (93.8)
 Pleural effusion
1 (3.1)
Sociodemographic variables, basal clinic, and molecular characteristics of both cohorts were taken from a centralized database (CLICaP Real World Data/Evidence database)
cfDNA circulating free DNA, CLICaP Latin American Consortium for Investigation of Lung Cancer, ND no data, TMB tumor mutational burden, TMB-H tumor mutation burden–high, TMB-L tumor mutation burden–low, ECOG Eastern Cooperative Oncology Group, NOS Non specified, PD-L1 Programmed Cell Death Ligand 1. 
The baseline TMB was obtained in 27 patients, among whom 22 (68.8%) had a low TMB (TMB-L). It is important to note that cohort A included five patients (31.3%) with TMB-H, while in cohort A, all patients with available data (11/16; 68.8%) had TMB-L (P = 0.054). Based on the mRNA levels stratification, there was a statistically significant difference when AXL and BIM levels were compared among both cohorts (81.3%, N = 13, cohort A patients had AXL-H; 93.8%, N = 15, had BIM-H; 75%, N = 12, cohort B patients had low AXL [AXL-L]; 85.7%, N = 14, had low BIM [BIM-L]). As shown in Supplementary Fig. 1, both differences in mRNA levels between cohorts A and B were statistically significant. Monitoring of response to osimertinib using ddPCR multiplex assays performed at week 8 was positive in 18 patients (56.3%); 93.8% and 18.8% of patients in cohorts A and B had cfDNA detectable by ddPCR, and this difference was statistically significant (P = 0.0001; Supplementary Fig. 2). In terms of PD-L1, most patients had < 1% (N = 3; 18.8%) and 1–49% (N = 11; 68.8%) in cohort A. Similarly, in cohort B, most patients had < 1% (N = 6; 37.5%) and 1–49% (N = 10; 62.5%). Only two patients had PD-L1 ≥ 50% in the study population, both from cohort A.

3.2 Outcomes Obtained with First-Line Osimertinib

The median time between the first sample and the start of therapy was 15 days, and the median time between the start of therapy and the first follow-up evaluation was 60 days (± 4 days). The ORRs for cohorts A and B were 6.3% and 100.0% (P = 0.0001), respectively. Only one of the patients with primary resistance to osimertinib had a short-lived PR, with subsequent multifocal progression, including the brain. On the contrary, in cohort B, 31.3% achieved a CR and 68.8% a PR. In both cohorts, the leading site of progression was the lung (≈60%), and cohort A patients had twice as many brain metastases, generally in the first 6 months after diagnosis (P = 0.67). PFS with osimertinib was 3.1 months (95% CI 0.47–7.0) for cohort A and NR for cohort B (P = 0.0001) (Fig. 3A). PFS was significantly higher in patients with ex19del compared with those with L858R mutations (24.5 months, 95% CI 18.2–NR, vs. 7.6 months, 95% CI 4.8–21.1; P = 0.001) (Supplementary Fig. 3; see the electronic supplementary material) and in those without brain metastases at diagnosis compared with patients with baseline brain metastases (24.5 months, 95% CI 18.5–30.5, vs. 7.2 months, 95% CI 1.1–13.2; P = 0.004) (Supplementary Fig. 4). The baseline T790M mutation also negatively impacted PFS (Supplementary Fig. 5); similarly, the presence of mutations in TP53 (P = 0.001), a higher number of commutations at diagnosis (P = 0.0001; Supplementary Fig. 6), and TMB-H (P = 0.017; Supplementary Fig. 7) were associated with worse PFS. In contrast, the PD-L1 expression level did not significantly affect PFS (P = 0.36). Given the biological differences between cohort A and B, high mRNA levels of AXL (P = 0.0001) and low BIM (P = 0.001) also affected PFS, as did the presence of cfDNA in liquid biopsies obtained 8 weeks after starting treatment with osimertinib (P = 0.001).
After a median follow-up of 25.1 months (95% CI 11.0–43.0), the OS for cohorts A and B was 20.1 months (95% CI 10.4–29.2) and NR (P = 0.0001, Fig. 3B). OS was higher for patients with ex19del (P = 0.002; Supplementary Fig. 3B), those without baseline brain metastases (P = 0.013; Supplementary Fig. 4B), those with two or fewer commutations (P = 0001; Supplementary Fig. 6B), and those with TMB-L (P = 0.0001; Supplementary Fig. 7B).
PD-L1 expression was significantly associated with OS, favoring the group of patients with a negative immunophenotype (PD-L1 < 1% 34.1 months, 95% CI 30.6–37.6, vs. PD-L1 1–49% 31.6 months, 95% CI 13.7–24.5, vs. PD-L1 ≥ 50% 19.1 months, 95% CI 13.7–24.5; P = 0.029). Low mRNA expression levels for AXL (P = 0.001) and high mRNA expression levels for BIM (P = 0.0001) and the absence of detectable cfDNA at week 8 of treatment (P = 0.001) were also significantly associated with OS (Supplementary Fig. 8A–C). Contrary to the findings for PFS, the presence of baseline T790M did not significantly impact OS (P = 0.06; Supplementary Fig. 5B). Univariate analysis showed that the variables that most negatively influenced PFS and OS were the presence of brain metastases (P = 0.002), evidence of a more significant number of commutations (P = 0.001), TMB-H (P = 0.0001), and the high levels of mRNA of AXL (P = 0.01) and low BIM (P = 0.02).

3.3 Genomic Alterations Found in Liquid Biopsy After Progression to Osimertinib

Figure 4 summarizes the main alterations in the liquid biopsy after progression to osimertinib (31 patients had a liquid biopsy, and one patient had pleural effusion cell block analysis). Sixty-four genomic alterations were found at the time of progression, 45 in cohort A (70%) and 19 in cohort B (30%). In cohort A, six patients (37.5%) lost the EGFR-sensitizing mutation, a condition that did not occur in any patient in cohort B (P = 0.01). In addition, in cohort A, off-target alterations were dominant, mainly in TP53 (13/35.5%), the RAS pathway (13/35.5%), PI3K (4/8.8%), and RB1 (2/4.4%). In contrast, most alterations in cohort B were due to on-target (6/31.2%) or included MET amplification (3/15.8%), the RAS pathway (2/10.5%), GNAS (2/ 10.5%), or RB1 (2/10.5%). After the second line, two patients had transformation to small cell carcinoma (6.3%), confirmed by biopsy, with mutations in TP53 and RB1. Post-osimertinib mean TMB was 5.0 (SD ± 3.6), with no significant differences between the two cohorts. Supplementary Fig. 9 shows a representative case of primary resistance to osimertinib, including genomic variations throughout the history of the disease (see the electronic supplementary material).

3.4 Second-Line Therapy

Thirty-one patients received second-line treatment (95.6%), mainly with the IMPOWER-150 study schedule (75%) or with the carboplatin/pemetrexed/bevacizumab combination (12.5%). The ORR for the second line was 68.8% (23 cases achieved PR), with no statistically significant difference between the two cohorts. The median PFS obtained with the second line was 8.1 months (95% CI 3.2–12.1) and 11.2 months (95% CI 9.8–12.5; P = 0.22) for cohorts A and B, respectively. The only variable that affected PFS for the second line was PD-L1 expression (PD-L1 < 1% 7.8 months, 95% CI 6.4–8.7, vs. PD-L1 1–49% 9.2 months, 95% CI 8.7–11.2, vs. PD-L1 ≥ 50% 11.6 months, 95% CI 7.3–13.7; P = 0.021).

4 Discussion

The mutational landscape for EGFR-TKI-sensitizing alterations plays a significant role in determining treatment efficacy for patients with NSCLC, and an in-depth understanding of this landscape offers an improved therapeutic approach. Nonetheless, the alterations that involve primary resistance to treatment with osimertinib among patients with EGFRm NSCLC have not been extensively characterized according to the observed ethnic variation in low-middle-income countries. In this study, we characterized the molecular determinants present in tumors of patients who do not respond to first-line treatment with osimertinib and compared them to patients with durable responses to osimertinib. Our results demonstrated that potential factors associated with poor survival outcomes under osimertinib treatment include the presence of three or more commutations (mainly including the TP53 gene), high levels of AXL mRNA, low levels of BIM mRNA, T790M gene de novo presence, TMB-H, and EGFR p.L858R variant presence.
Although TKI-acquired resistance has been extensively studied, there are more studies about the molecular interactions that bypass the action of TKI than primary resistance [34]. After exposure to osimertinib, ORR was considerably lower in cohort A (6.3%), with lung involvement as the leading site of progression and two times higher brain involvement. EGFRm are associated with the progression of the disease, and the brain is one of the primary sites of metastasis; in fact, patients with T790M usually have a higher brain compromise [35].
TMB-H, high expression levels of AXL mRNA, and low mRNA levels of BIM were commonly identified in patients with intrinsic resistance. These alterations could add genomic instability burden to the tumor and, in fact, resistance to TKI inhibitors [36, 37]. AXL is a tyrosine kinase receptor that transduces signals from the extracellular matrix into the cytoplasm by binding growth factor GAS6, thus regulating many physiological processes, including cell survival, cell proliferation, migration, and differentiation [38]. High expression of the AXL protein in tumors is reported to be associated with poor prognosis and acquired resistance to targeted therapy in several types of cancer, including glioblastoma, breast cancer, lung cancer, and acute myeloid leukemia [39, 40]. Recently, Taniguchi et al. showed that osimertinib stimulated AXL, which binds to the HER family proteins EGFR and HER3 through a negative feedback loop involving the suppression of sprouty RTK signaling antagonist 4 (SPRY4). In sequence, activated AXL was associated with EGFR and HER3 in maintaining cell survival and inducing the emergence of cells tolerant to osimertinib [36]. Likewise, AXL hyperactivity also includes resistance through EMT closely associated with tumor heterogeneity and induces variations in pathways, including MET, platelet-derived growth factor receptor (PDGFR), and MAPK [39, 41, 42]. In our study, 81% of the cases in cohort A had high levels of AXL mRNA expression, values that were 1.5 times higher than those observed in cohort B.
As mentioned above, low levels of BIM mRNA were associated with a worse prognosis. BIM is a BH3-only protein that directly activates the ultimate effectors of apoptosis BAK (BCL-2 antagonist or killer) and BAX (BCL-2-associated X protein) [43]. Recently, Karachaliou and colleagues assessed the role of BIM mRNA expression in patients treated in the EURTAC study [44]. The analysis shows that PFS and OS were significantly longer for patients with high BIM expression than for those with low/intermediate BIM mRNA expression. Patients with low BIM expression could derive only a meager benefit from treatment with EGFR-TKI alone. Still, they could benefit from synthetic lethality combinations, including small molecules that mimic the BH3 motif [30]. Previous evidence suggested that gefitinib combined with the BH3 mimetic ABT-737 (an analog of navitoclax) substantially increases apoptosis compared with each agent alone in EGFR-mutant H1650 cells with low BIM expression [37, 45].
PFS and OS were also significantly lower among patients with primary resistance. In cohort A, patients with ex19del had a higher PFS (and presence of brain metastasis) than those with L858R. Hong et al. had previously described that L858R could have a negative prognostic value in NSCLC patients. In their cohort, patients with mutations in exon 21 had a PFS of almost 8 months [4648]. In a similar way, in a Korean cohort of patients, ex19del and no brain involvement were associated with a better PFS under TKI treatment [49]. However, for the same cohort, PFS was negatively affected by the presence of T790M, even under treatment with third-line TKI osimertinib. This finding could be explained under the theory of T790M as one possible intrinsic resistance mechanism. However, T790M can also favor other resistance mechanisms, MET amplification among them [50]. In our study, cohort A reported a big proportion of basal L858R mutation, whereas cohort B had ex19del predominately. It is essential to address that the presence of ex19del has been associated with better outcomes [3, 8]. However, commutations alter disease progression by modulating the sensitivity to TKI and facilitating more aggressive tumor biology. ex19del alterations are associated with TMB-L impacting OS and not PFS. As Choi et al. demonstrated, the presence of ex19del is associated with a good OS, even under treatment with first-line TKI [51]. However, in our study, patients were treated with a third-generation TKI with better efficacy than other first- or second-generation TKI. In the absence of L858R mutation, the better outcomes under TKI could be related to the description of EGFR ex19del as a more sensitizing mutation that causes changes in phosphorylation and downstream signaling that result in a better response to osimertinib [52, 53].
TMB has been characterized as a possible predictor factor of targeted therapy response. Previous studies have reported the association between TMB-H with outcomes and a shorter time to treatment discontinuation among patients with EGFR-mutated NSCLC under TKI treatment. A cohort presented by Offin et al. showed a negative relationship between a higher TMB and OS [54]. Some authors mention that TMB-H can be associated with the sub-clone presence that could favor resistant cell populations under TKI treatment. TMB could be a marker of propensity towards mutagenesis, mainly among EGFR-mutant NSCLC [55, 56]. Interestingly, after osimertinib treatment, TMB was not different between the cohorts, suggesting that genomic heterogenicity could be pre-established before selection under TKI.
Regarding the genomic alterations presented after progression to osimertinib, almost two-thirds were identified among patients in the cohort classified as having primary resistance. For this group, some patients lost the EGFR-sensitizing mutation. In vitro studies have suggested that this loss could be a possible mechanism of TKI resistance, at least for first- or second-generation TKI [57]. Remarkably, for the cohort with primary resistance, the genomic alterations identified were off-target mutations. TP53 and KRAS pathway RB1 were the most common. In the context of the EGFR-independent resistance mechanism, TP53 and RB1 tumor suppressor genes could be identified in almost 50% of LUAD among Caucasian patients [26, 58, 59]. According to the evidence, TP53 alterations appear during advanced disease, playing a key role in tumor progression more than tumor initiation. TP53/RB1 mutations in NSCLC are associated with transformation to SCLC under TKI treatment [60, 61]. The two patients who presented phenotypic transformation had TP53 and RB1 mutations at the time of biopsy confirmation. KRAS is another EGFR-independent resistance mechanism that was reported in our study. Although EGFR and KRAS commutations are rare, KRAS amplification could favor resistance through ERBB-family upregulation [20, 62] As expected, in cohort B, MET amplification was a common mutation. It is known that under treatment with TKI, MET alterations can be found in almost 5% and 20% of patients treated with first- and second-generation EGFR-TKI and osimertinib, respectively [23, 63, 64].
This study, as with other studies focused on specific genomic characterization of primary resistance, has some limitations, such as the small sample size and the specific population that affects external validity. Also, based on the characteristics of the patients, matching both cohorts was not feasible, which might limit the extent of the comparison between cohorts.

5 Conclusions

Understanding the molecular mechanisms that favor primary resistance to EGFR-TKI is fundamental to choosing NSCLC treatment and finding new molecular targets. Targeting EGFR could force the selection of tumors with other molecular pathway alterations that allow tumor progression. EGFR-independent mechanisms of resistance (or off-target alterations) were found in a population with primary resistance. Treatment with the third-generation TKI osimertinib produces very limited survival outcomes for patients with off-target alterations. These data evidence the need for molecular profiling of patients before the beginning of treatment and the challenge of developing new therapeutic approaches based on specific molecular alterations. As the presence of cfDNA at the time of progression was associated with poor outcomes, liquid biopsy reinforces its role in monitoring the prognosis among NSCLC Hispanic patients.

Acknowledgements

Dora Lucia Vallejo-Ardila is supported by the Department of Science, Technology, and Innovation (COLCIENCIAS-Colombia) as part of her PhD overseas 679 scholarship program.

Declarations

Funding

This study was sponsored by the stipend PCI-2018-10171 of Universidad El Bosque (Bogotá, Colombia) and approved by the Institutional Ethics Committee (reference No. 019-2021). The CLICaP and the Foundation for Clinical and Applied Cancer Research—FICMAC (Bogotá, Colombia) supported some lab experiments through research grant 023-2019.

Conflict of interest

Andrés F. Cardona discloses financial research support from Merck Sharp & Dohme, Boehringer Ingelheim, Roche, Bristol-Myers Squibb, and the Foundation for Clinical and Applied Cancer Research—FICMAC. Additionally, he was linked to and received honoraria as an advisor to, participated in speakers' bureau for, and gave expert testimony to Merck Sharp & Dohme, Boehringer Ingelheim, Roche, Bristol-Myers Squibb, Pfizer, Novartis, Celldex Therapeutics, Foundation Medicine, Eli Lilly, and the Foundation for Clinical and Applied Cancer Research—FICMAC. Oscar Arrieta reports personal fees from Pfizer, grants and individual fees from Astra Zeneca, grants and individual fees from Boehringer-Ingelheim, personal fees from Lilly, individual fees from Merck, personal fees from Bristol Myers Squibb, and grants and personal fees from Roche, outside the submitted work. Gonzalo Recondo reports personal fees from Pfizer, Roche, Amgen, Bayer, Takeda, MSD, and Merck Serono, and research grants from Amgen and Janssen, outside of the submitted work. Juan Blaquier has nothing to disclose.

Ethics approval

All included patients provided signed informed consent. In addition, an Institutional Review Board and Privacy Board waiver was obtained to facilitate retrospective clinical-pathological and molecular data (Lung Cancer-FICMAC/CLICaP Platform—Registration No. 2012/014, Kayre, Bogotá, Colombia, and Research Ethics Committee of Universidad El Bosque—Registration No. PCI-2018-10171).
See the ethics approval statement.
Not applicable.

Code availability

Not applicable.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat O’Leary C, Gasper H, Sahin KB, Tang M, Kulasinghe A, Adams MN, et al. Epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer (NSCLC). Pharmaceuticals. 2020;13(10):1–16.CrossRef O’Leary C, Gasper H, Sahin KB, Tang M, Kulasinghe A, Adams MN, et al. Epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer (NSCLC). Pharmaceuticals. 2020;13(10):1–16.CrossRef
2.
Zurück zum Zitat Piotrowska Z, Sequist LV. Epidermal growth factor receptor-mutant lung cancer new drugs, new resistance mechanisms, and future treatment options. Cancer J. 2015;21(5):371–2.PubMedCrossRef Piotrowska Z, Sequist LV. Epidermal growth factor receptor-mutant lung cancer new drugs, new resistance mechanisms, and future treatment options. Cancer J. 2015;21(5):371–2.PubMedCrossRef
3.
Zurück zum Zitat Midha A, Dearden S, Mccormack R. EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity (mutMapII). Am J Cancer Res. 2015;5(9):2892–911.PubMedPubMedCentral Midha A, Dearden S, Mccormack R. EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity (mutMapII). Am J Cancer Res. 2015;5(9):2892–911.PubMedPubMedCentral
4.
Zurück zum Zitat Campbell JD, Lathan C, Sholl L, Ducar M, Vega M, Sunkavalli A, et al. Comparison of prevalence and types of mutations in lung cancers among black and white populations. JAMA Oncol. 2017;3(6):801–9.PubMedPubMedCentralCrossRef Campbell JD, Lathan C, Sholl L, Ducar M, Vega M, Sunkavalli A, et al. Comparison of prevalence and types of mutations in lung cancers among black and white populations. JAMA Oncol. 2017;3(6):801–9.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Arrieta O, Cardona AF, Martín C, Más-López L, Corrales-Rodríguez L, Bramuglia G, et al. Updated frequency of EGFR and KRAS mutations in NonSmall-cell lung cancer in Latin America: the Latin-American consortium for the investigation of lung cancer (CLICaP). J Thorac Oncol. 2015;10(5):838–43.PubMedCrossRef Arrieta O, Cardona AF, Martín C, Más-López L, Corrales-Rodríguez L, Bramuglia G, et al. Updated frequency of EGFR and KRAS mutations in NonSmall-cell lung cancer in Latin America: the Latin-American consortium for the investigation of lung cancer (CLICaP). J Thorac Oncol. 2015;10(5):838–43.PubMedCrossRef
6.
Zurück zum Zitat Gimbrone NT, Sarcar B, Gordian ER, Rivera JI, Lopez C, Yoder SJ, et al. Somatic mutations and ancestry markers in hispanic lung cancer patients. J Thorac Oncol. 2017;12(12):1851–6.PubMedPubMedCentralCrossRef Gimbrone NT, Sarcar B, Gordian ER, Rivera JI, Lopez C, Yoder SJ, et al. Somatic mutations and ancestry markers in hispanic lung cancer patients. J Thorac Oncol. 2017;12(12):1851–6.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Leal LF, de Paula FE, de Marchi P, de Souza VL, Pinto GDJ, Carlos CD, et al. Mutational profile of Brazilian lung adenocarcinoma unveils association of EGFR mutations with high Asian ancestry and independent prognostic role of KRAS mutations. Sci Rep. 2019;9(1):3209.PubMedPubMedCentralCrossRef Leal LF, de Paula FE, de Marchi P, de Souza VL, Pinto GDJ, Carlos CD, et al. Mutational profile of Brazilian lung adenocarcinoma unveils association of EGFR mutations with high Asian ancestry and independent prognostic role of KRAS mutations. Sci Rep. 2019;9(1):3209.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Chen J, Yang H, Teo ASM, Amer LB, Sherbaf FG, Tan CQ, et al. Genomic landscape of lung adenocarcinoma in East Asians. Nat Genet. 2020;52(2):177–86.PubMedCrossRef Chen J, Yang H, Teo ASM, Amer LB, Sherbaf FG, Tan CQ, et al. Genomic landscape of lung adenocarcinoma in East Asians. Nat Genet. 2020;52(2):177–86.PubMedCrossRef
9.
Zurück zum Zitat Koivunen JP, Kim J, Lee J, Rogers AM, Park JO, Zhao X, et al. Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients. Br J Cancer. 2008;99(2):245–52.PubMedPubMedCentralCrossRef Koivunen JP, Kim J, Lee J, Rogers AM, Park JO, Zhao X, et al. Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients. Br J Cancer. 2008;99(2):245–52.PubMedPubMedCentralCrossRef
10.
11.
Zurück zum Zitat Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl Med. 2004;21(20):2129–39.CrossRef Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl Med. 2004;21(20):2129–39.CrossRef
12.
Zurück zum Zitat Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169–81.PubMedCrossRef Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169–81.PubMedCrossRef
13.
Zurück zum Zitat D’Angelo SP, Pietanza MC, Johnson ML, Riely GJ, Miller VA, Sima CS, et al. Incidence of EGFR Exon 19 deletions and 1858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol. 2011;29(15):2066–70.PubMedPubMedCentralCrossRef D’Angelo SP, Pietanza MC, Johnson ML, Riely GJ, Miller VA, Sima CS, et al. Incidence of EGFR Exon 19 deletions and 1858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol. 2011;29(15):2066–70.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat 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(2):113–25.PubMedCrossRef 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(2):113–25.PubMedCrossRef
16.
Zurück zum Zitat Ramalingam SS, Yang C-HJ, Khoon Lee C, Kurata T, Kim DW, John T, et al. Osimertinib as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer. J Clin Oncol. 2017;36:841–9.PubMedCrossRef Ramalingam SS, Yang C-HJ, Khoon Lee C, Kurata T, Kim DW, John T, et al. Osimertinib as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer. J Clin Oncol. 2017;36:841–9.PubMedCrossRef
17.
Zurück zum Zitat Reungwetwattana T, Nakagawa K, Cho BC, Cobo M, Cho EK, Bertolini A, 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;36:3290–7.CrossRef Reungwetwattana T, Nakagawa K, Cho BC, Cobo M, Cho EK, Bertolini A, 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;36:3290–7.CrossRef
18.
Zurück zum Zitat Lorenzi M, Ferro A, Cecere F, Scattolin D, del Conte A, Follador A, et al. First-line osimertinib in patients with EGFR-mutant advanced non-small cell lung cancer: outcome and safety in the real world: FLOWER study. Oncologist. 2021;27(2):E87-115.CrossRef Lorenzi M, Ferro A, Cecere F, Scattolin D, del Conte A, Follador A, et al. First-line osimertinib in patients with EGFR-mutant advanced non-small cell lung cancer: outcome and safety in the real world: FLOWER study. Oncologist. 2021;27(2):E87-115.CrossRef
19.
Zurück zum Zitat Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Jänne PA, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol. 2010;28(2):357–60.PubMedCrossRef Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Jänne PA, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol. 2010;28(2):357–60.PubMedCrossRef
20.
Zurück zum Zitat Wang J, Wang B, Chu H, Yao Y. Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFR mutations. OncoTargets Therapy. 2016;9:3711–26.PubMedPubMedCentralCrossRef Wang J, Wang B, Chu H, Yao Y. Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFR mutations. OncoTargets Therapy. 2016;9:3711–26.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Passaro A, Jänne PA, Mok T, Peters S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat Cancer. 2021;2:377–91.PubMedCrossRef Passaro A, Jänne PA, Mok T, Peters S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat Cancer. 2021;2:377–91.PubMedCrossRef
22.
Zurück zum Zitat Dong L, Lei D, Zhang H. Clinical strategies for acquired epidermal growth factor receptor tyrosine kinase inhibitor resistance in non-small-cell lung cancer patients. Oncotarget. 2017;8(38):64600–6.PubMedPubMedCentralCrossRef Dong L, Lei D, Zhang H. Clinical strategies for acquired epidermal growth factor receptor tyrosine kinase inhibitor resistance in non-small-cell lung cancer patients. Oncotarget. 2017;8(38):64600–6.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Minari R, Bordi P, Tiseo M. Third-generation epidermal growth factor receptor-tyrosine kinase inhibitors in T790M-positive non-small cell lung cancer: review on emerged mechanisms of resistance. Transl Lung Cancer Res. 2016;5(6):695–708.PubMedPubMedCentralCrossRef Minari R, Bordi P, Tiseo M. Third-generation epidermal growth factor receptor-tyrosine kinase inhibitors in T790M-positive non-small cell lung cancer: review on emerged mechanisms of resistance. Transl Lung Cancer Res. 2016;5(6):695–708.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Blakely CM, Watkins TBK, Wu W, Gini B, Chabon JJ, McCoach CE, et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat Genet. 2017;49(12):1693–704.PubMedPubMedCentralCrossRef Blakely CM, Watkins TBK, Wu W, Gini B, Chabon JJ, McCoach CE, et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat Genet. 2017;49(12):1693–704.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Jordan EJ, Kim HR, Arcila ME, Barron D, Chakravarty D, Gao JJ, et al. Prospective comprehensive molecular characterization of lung adenocarcinomas for efficient patient matching to approved and emerging therapies. Cancer Discov. 2017;7(6):596–609.PubMedPubMedCentralCrossRef Jordan EJ, Kim HR, Arcila ME, Barron D, Chakravarty D, Gao JJ, et al. Prospective comprehensive molecular characterization of lung adenocarcinomas for efficient patient matching to approved and emerging therapies. Cancer Discov. 2017;7(6):596–609.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Santoni-Rugiu E, Melchior LC, Urbanska EM, Jakobsen JN, de Stricker K, Grauslund M, et al. Intrinsic resistance to EGFR-tyrosine kinase inhibitors in EGFR-mutant non-small cell lung cancer: differences and similarities with acquired resistance. Cancers (Basel). 2019;11(7):923.PubMedPubMedCentralCrossRef Santoni-Rugiu E, Melchior LC, Urbanska EM, Jakobsen JN, de Stricker K, Grauslund M, et al. Intrinsic resistance to EGFR-tyrosine kinase inhibitors in EGFR-mutant non-small cell lung cancer: differences and similarities with acquired resistance. Cancers (Basel). 2019;11(7):923.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O’Connell A, Messineo MM, et al. Noninvasive detection of response and resistance in egfrmutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res. 2014;20(6):1698–705.PubMedPubMedCentralCrossRef Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O’Connell A, Messineo MM, et al. Noninvasive detection of response and resistance in egfrmutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res. 2014;20(6):1698–705.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Zhu G, Ye X, Dong Z, Lu YC, Sun Y, Liu Y, et al. Highly sensitive droplet digital PCR method for detection of EGFR-activating mutations in plasma cell-free DNA from patients with advanced non-small cell lung cancer. J Mol Diagn. 2015;17(3):265–72.PubMedCrossRef Zhu G, Ye X, Dong Z, Lu YC, Sun Y, Liu Y, et al. Highly sensitive droplet digital PCR method for detection of EGFR-activating mutations in plasma cell-free DNA from patients with advanced non-small cell lung cancer. J Mol Diagn. 2015;17(3):265–72.PubMedCrossRef
29.
Zurück zum Zitat Karachaliou N, Chaib I, Cardona AF, Berenguer J, Bracht JWP, Yang J, et al. Common co-activation of AXL and CDCP1 in EGFR-mutation-positive non-smallcell lung cancer associated with poor prognosis. EBioMedicine. 2018;29:112–27.PubMedPubMedCentralCrossRef Karachaliou N, Chaib I, Cardona AF, Berenguer J, Bracht JWP, Yang J, et al. Common co-activation of AXL and CDCP1 in EGFR-mutation-positive non-smallcell lung cancer associated with poor prognosis. EBioMedicine. 2018;29:112–27.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Karachaliou N, Codony-Servat J, Teixidó C, Pilotto S, Drozdowskyj A, Codony-Servat C, et al. BIM and mTOR expression levels predict outcome to erlotinib in EGFR-mutant non-small-cell lung cancer. Sci Rep. 2015;5:17499.PubMedPubMedCentralCrossRef Karachaliou N, Codony-Servat J, Teixidó C, Pilotto S, Drozdowskyj A, Codony-Servat C, et al. BIM and mTOR expression levels predict outcome to erlotinib in EGFR-mutant non-small-cell lung cancer. Sci Rep. 2015;5:17499.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Marabelle A, Le DT, Ascierto PA, di Giacomo M, de Jesus-Acosta A, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/ mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38(1):1–10.PubMedCrossRef Marabelle A, Le DT, Ascierto PA, di Giacomo M, de Jesus-Acosta A, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/ mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38(1):1–10.PubMedCrossRef
32.
Zurück zum Zitat Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14(2):178–92.PubMedCrossRef Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14(2):178–92.PubMedCrossRef
33.
Zurück zum Zitat Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24–6.PubMedPubMedCentralCrossRef Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24–6.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Nan X, Xie C, Yu X, Liu J. EGFR TKI as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer. Oncotarget. 2017;8(43):75712–26.PubMedPubMedCentralCrossRef Nan X, Xie C, Yu X, Liu J. EGFR TKI as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer. Oncotarget. 2017;8(43):75712–26.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Chen Y, Deng J, Liu Y, Wang H, Zhao S, He Y, et al. Analysis of metastases in non-small cell lung cancer patients with epidermal growth factor receptor mutation. Ann Transl Med. 2021;9(3):206–206.PubMedPubMedCentralCrossRef Chen Y, Deng J, Liu Y, Wang H, Zhao S, He Y, et al. Analysis of metastases in non-small cell lung cancer patients with epidermal growth factor receptor mutation. Ann Transl Med. 2021;9(3):206–206.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Taniguchi H, Yamada T, Wang R, Tanimura K, Adachi Y, Nishiyama A, et al. AXL confers intrinsic resistance to osimertinib and advances the emergence of tolerant cells. Nat Commun. 2019;10(1):259.CrossRefPubMedPubMedCentral Taniguchi H, Yamada T, Wang R, Tanimura K, Adachi Y, Nishiyama A, et al. AXL confers intrinsic resistance to osimertinib and advances the emergence of tolerant cells. Nat Commun. 2019;10(1):259.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Su W, Zhang X, Cai X, Peng M, Wang F, Wang Y. BIM deletion polymorphism predicts poor response to EGFR-TKIs in nonsmall cell lung cancer: An updated meta-analysis. Medicine. 2019;98(10): e14568.PubMedPubMedCentralCrossRef Su W, Zhang X, Cai X, Peng M, Wang F, Wang Y. BIM deletion polymorphism predicts poor response to EGFR-TKIs in nonsmall cell lung cancer: An updated meta-analysis. Medicine. 2019;98(10): e14568.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Zhu C, Wei Y, Wei X. AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications. Mol Cancer. 2019;18(1):153.PubMedPubMedCentralCrossRef Zhu C, Wei Y, Wei X. AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications. Mol Cancer. 2019;18(1):153.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Zhang Z, Lee JC, Lin L, Olivas V, Au V, Laframboise T, et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet. 2012;44(8):852–60.PubMedPubMedCentralCrossRef Zhang Z, Lee JC, Lin L, Olivas V, Au V, Laframboise T, et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet. 2012;44(8):852–60.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wu Z, Bai F, Fan L, Pang W, Han R, Wang J, et al. Coexpression of receptor tyrosine kinase AXL and EGFR in human primary lung adenocarcinomas. Hum Pathol. 2015;46(12):1935–44.PubMedCrossRef Wu Z, Bai F, Fan L, Pang W, Han R, Wang J, et al. Coexpression of receptor tyrosine kinase AXL and EGFR in human primary lung adenocarcinomas. Hum Pathol. 2015;46(12):1935–44.PubMedCrossRef
41.
Zurück zum Zitat Brand TM, Iida M, Stein AP, Corrigan KL, Braverman CM, Luthar N, et al. AXL mediates resistance to cetuximab therapy. Cancer Res. 2014;74(18):5152–64.PubMedPubMedCentralCrossRef Brand TM, Iida M, Stein AP, Corrigan KL, Braverman CM, Luthar N, et al. AXL mediates resistance to cetuximab therapy. Cancer Res. 2014;74(18):5152–64.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Meyer AS, Miller MA, Gertler FB, Lauffenburger DA. The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci Signal. 2013;6(287):ra66.CrossRefPubMedPubMedCentral Meyer AS, Miller MA, Gertler FB, Lauffenburger DA. The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci Signal. 2013;6(287):ra66.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Letai A, Bassik M, Walensky L, Sorcinelli M, Weiler S, Korsmeyer S. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2(3):183–92.PubMedCrossRef Letai A, Bassik M, Walensky L, Sorcinelli M, Weiler S, Korsmeyer S. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2(3):183–92.PubMedCrossRef
44.
Zurück zum Zitat Costa C, Molina MA, Drozdowskyj A, Gimeńez-Capitán A, Bertran-Alamillo J, Karachaliou N, et al. The impact of EGFR T790M mutations and BIM mRNA expression on outcome in patients with EGFR-Mutant NSCLC treated with erlotinib or chemotherapy in the randomized phase III EURTAC trial. Clin Cancer Res. 2014;20(7):2001–10.PubMedCrossRef Costa C, Molina MA, Drozdowskyj A, Gimeńez-Capitán A, Bertran-Alamillo J, Karachaliou N, et al. The impact of EGFR T790M mutations and BIM mRNA expression on outcome in patients with EGFR-Mutant NSCLC treated with erlotinib or chemotherapy in the randomized phase III EURTAC trial. Clin Cancer Res. 2014;20(7):2001–10.PubMedCrossRef
45.
Zurück zum Zitat Cragg M, Kuroda J, Puthalakath H, Huang D, Strasser A. Gefitinib-induced killing of NSCLC cell lines expressing mutant EGFR requires BIM and can be enhanced by BH3 mimetics. PLoS ONE. 2007;4(10):1681–9. Cragg M, Kuroda J, Puthalakath H, Huang D, Strasser A. Gefitinib-induced killing of NSCLC cell lines expressing mutant EGFR requires BIM and can be enhanced by BH3 mimetics. PLoS ONE. 2007;4(10):1681–9.
46.
Zurück zum Zitat Hong W, Wu Q, Zhang J, Zhou Y. Prognostic value of EGFR 19-del and 21–L858R mutations in patients with non-small cell lung cancer. Oncol Lett. 2019;18(4):3887–95.PubMedPubMedCentral Hong W, Wu Q, Zhang J, Zhou Y. Prognostic value of EGFR 19-del and 21–L858R mutations in patients with non-small cell lung cancer. Oncol Lett. 2019;18(4):3887–95.PubMedPubMedCentral
47.
Zurück zum Zitat Kuan FC, Kuo LT, Chen MC, Yang CT, Shi CS, Teng D, et al. Overall survival benefits of first-line EGFR tyrosine kinase inhibitors in EGFR-mutated non-small-cell lung cancers: a systematic review and meta-analysis. Br J Cancer. 2015;113(10):1519–28.PubMedPubMedCentralCrossRef Kuan FC, Kuo LT, Chen MC, Yang CT, Shi CS, Teng D, et al. Overall survival benefits of first-line EGFR tyrosine kinase inhibitors in EGFR-mutated non-small-cell lung cancers: a systematic review and meta-analysis. Br J Cancer. 2015;113(10):1519–28.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Leal JL, Alexander M, Itchins M, Wright GM, Kao S, Hughes BGM, et al. EGFR Exon 20 insertion mutations: clinicopathological characteristics and treatment outcomes in advanced non-small cell lung cancer: EGFR Exon 20 insertion mutations in NSCLC. Clin Lung Cancer. 2021;22(6):e859–69.PubMedCrossRef Leal JL, Alexander M, Itchins M, Wright GM, Kao S, Hughes BGM, et al. EGFR Exon 20 insertion mutations: clinicopathological characteristics and treatment outcomes in advanced non-small cell lung cancer: EGFR Exon 20 insertion mutations in NSCLC. Clin Lung Cancer. 2021;22(6):e859–69.PubMedCrossRef
49.
Zurück zum Zitat Jung HA, Woo SY, Lee SH, Ahn JS, Ahn MJ, Park K, et al. The different central nervous system efficacy among gefitinib, erlotinib and afatinib in patients with epidermal growth factor receptor mutation-positive non-small cell lung cancer. Transl Lung Cancer Res. 2020;9(5):1749–58.PubMedPubMedCentralCrossRef Jung HA, Woo SY, Lee SH, Ahn JS, Ahn MJ, Park K, et al. The different central nervous system efficacy among gefitinib, erlotinib and afatinib in patients with epidermal growth factor receptor mutation-positive non-small cell lung cancer. Transl Lung Cancer Res. 2020;9(5):1749–58.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Suda K, Murakami I, Katayama T, Tomizawa K, Osada H, Sekido Y, et al. Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer. Clin Cancer Res. 2010;16(22):5489–98.PubMedCrossRef Suda K, Murakami I, Katayama T, Tomizawa K, Osada H, Sekido Y, et al. Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer. Clin Cancer Res. 2010;16(22):5489–98.PubMedCrossRef
51.
Zurück zum Zitat Choi YW, Jeon SY, Jeong GS, Lee HW, Jeong SH, Kang SY, et al. EGFR Exon 19 deletion is associated with favorable overall survival after first-line gefitinib therapy in advanced non-small cell lung cancer patients. Am J Clin Oncol. 2018;41(4):385–90.PubMedCrossRef Choi YW, Jeon SY, Jeong GS, Lee HW, Jeong SH, Kang SY, et al. EGFR Exon 19 deletion is associated with favorable overall survival after first-line gefitinib therapy in advanced non-small cell lung cancer patients. Am J Clin Oncol. 2018;41(4):385–90.PubMedCrossRef
52.
Zurück zum Zitat Okabe T, Okamoto I, Tamura K, Terashima M, Yoshida T, Satoh T, et al. Differential constitutive activation of the epidermal growth factor receptor in non-small cell lung cancer cells bearing EGFR gene mutation and amplification. Cancer Res. 2007;67(5):2046–53.PubMedCrossRef Okabe T, Okamoto I, Tamura K, Terashima M, Yoshida T, Satoh T, et al. Differential constitutive activation of the epidermal growth factor receptor in non-small cell lung cancer cells bearing EGFR gene mutation and amplification. Cancer Res. 2007;67(5):2046–53.PubMedCrossRef
53.
Zurück zum Zitat Zhu J, Zhong W, Zhang G, Li R, Zhang X, Guo A, et al. Better survival with EGFR exon 19 than exon 21 mutations in gefitinib-treated non-small cell lung cancer patients is due to differential inhibition of downstream signals. Cancer Lett. 2008;265(2):307–17.PubMedCrossRef Zhu J, Zhong W, Zhang G, Li R, Zhang X, Guo A, et al. Better survival with EGFR exon 19 than exon 21 mutations in gefitinib-treated non-small cell lung cancer patients is due to differential inhibition of downstream signals. Cancer Lett. 2008;265(2):307–17.PubMedCrossRef
54.
Zurück zum Zitat Offin M, Rizvi H, Tenet M, Ni A, Sanchez-Vega F, Li BT, et al. Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin Cancer Res. 2019;25(3):1063–9.PubMedCrossRef Offin M, Rizvi H, Tenet M, Ni A, Sanchez-Vega F, Li BT, et al. Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin Cancer Res. 2019;25(3):1063–9.PubMedCrossRef
55.
Zurück zum Zitat Cheng ML, Oxnard GR. Does TMB impact the effectiveness of TKIs in EGFR-mutant NSCLC? Clin Cancer Res. 2019;25(3):899–900.PubMedCrossRef Cheng ML, Oxnard GR. Does TMB impact the effectiveness of TKIs in EGFR-mutant NSCLC? Clin Cancer Res. 2019;25(3):899–900.PubMedCrossRef
56.
Zurück zum Zitat Lin C, Shi X, Zhao J, He Q, Fan Y, Xu W, et al. Tumor mutation burden correlates with efficacy of chemotherapy/targeted therapy in advanced non-small cell lung cancer. Front Oncol. 2020;10:480.PubMedPubMedCentralCrossRef Lin C, Shi X, Zhao J, He Q, Fan Y, Xu W, et al. Tumor mutation burden correlates with efficacy of chemotherapy/targeted therapy in advanced non-small cell lung cancer. Front Oncol. 2020;10:480.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Tabara K, Kanda R, Sonoda K, Kubo T, Murakami Y, Kawahara A, et al. Loss of activating EGFR mutant gene contributes to acquired resistance to EGFR tyrosine kinase inhibitors in lung cancer cells. PLoS ONE. 2012;7(7):e41017.PubMedPubMedCentralCrossRef Tabara K, Kanda R, Sonoda K, Kubo T, Murakami Y, Kawahara A, et al. Loss of activating EGFR mutant gene contributes to acquired resistance to EGFR tyrosine kinase inhibitors in lung cancer cells. PLoS ONE. 2012;7(7):e41017.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Jakobsen JN, Santoni-Rugiu E, Grauslund M, Melchior L, Sørensen JB. Concomitant driver mutations in advanced EGFR-mutated non-small-cell lung cancer and their impact on erlotinib treatment. Oncotarget. 2018;9(40):26195–208.PubMedPubMedCentralCrossRef Jakobsen JN, Santoni-Rugiu E, Grauslund M, Melchior L, Sørensen JB. Concomitant driver mutations in advanced EGFR-mutated non-small-cell lung cancer and their impact on erlotinib treatment. Oncotarget. 2018;9(40):26195–208.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Lee T, Lee B, la Choi Y, Han J, Ahn MJ, Um SW. Non-small cell lung cancer with concomitant EGFR, KRAS, and ALK mutation: clinicopathologic features of 12 cases. J Pathol Transl Med. 2016;50(3):197–203.PubMedPubMedCentralCrossRef Lee T, Lee B, la Choi Y, Han J, Ahn MJ, Um SW. Non-small cell lung cancer with concomitant EGFR, KRAS, and ALK mutation: clinicopathologic features of 12 cases. J Pathol Transl Med. 2016;50(3):197–203.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Marcoux N, Gettinger SN, Grainne O, Arbour KC, Neal JW, Husain H, et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. J Clin Oncol. 2019;37(4):278–85.PubMedCrossRef Marcoux N, Gettinger SN, Grainne O, Arbour KC, Neal JW, Husain H, et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. J Clin Oncol. 2019;37(4):278–85.PubMedCrossRef
61.
Zurück zum Zitat Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, et al. Tracking the evolution of non-small-cell lung cancer. N Engl J Med. 2017;376(22):2109–21.PubMedCrossRef Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, et al. Tracking the evolution of non-small-cell lung cancer. N Engl J Med. 2017;376(22):2109–21.PubMedCrossRef
62.
Zurück zum Zitat Stewart EL, Tan SZ, Liu G, Tsao MS. Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations-a review. Transl Lung Cancer Res. 2015;4(1):67–81.PubMedPubMedCentral Stewart EL, Tan SZ, Liu G, Tsao MS. Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations-a review. Transl Lung Cancer Res. 2015;4(1):67–81.PubMedPubMedCentral
63.
Zurück zum Zitat Piotrowska Z, Isozaki H, Lennerz JK, Gainor JF, Lennes IT, Zhu VW, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov. 2018;8(12):1529–39.PubMedPubMedCentralCrossRef Piotrowska Z, Isozaki H, Lennerz JK, Gainor JF, Lennes IT, Zhu VW, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov. 2018;8(12):1529–39.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Tetsu O, Hangauer MJ, Phuchareon J, Eisele DW, McCormick F. Drug resistance to EGFR inhibitors in lung cancer. Chemotherapy. 2016;61(5):223–35.PubMedCrossRef Tetsu O, Hangauer MJ, Phuchareon J, Eisele DW, McCormick F. Drug resistance to EGFR inhibitors in lung cancer. Chemotherapy. 2016;61(5):223–35.PubMedCrossRef
Metadaten
Titel
Genomic Landscape of Primary Resistance to Osimertinib Among Hispanic Patients with EGFR-Mutant Non-Small Cell Lung Cancer (NSCLC): Results of an Observational Longitudinal Cohort Study
verfasst von
Diego F. Chamorro
Andrés F. Cardona
July Rodríguez
Alejandro Ruiz-Patiño
Oscar Arrieta
Darwin A. Moreno-Pérez
Leonardo Rojas
Zyanya Lucia Zatarain-Barrón
Dora V. Ardila
Lucia Viola
Gonzalo Recondo
Juan B. Blaquier
Claudio Martín
Luis Raez
Suraj Samtani
Camila Ordóñez-Reyes
Juan Esteban Garcia-Robledo
Luis Corrales
Carolina Sotelo
Luisa Ricaurte
Mauricio Cuello
Sergio Mejía
Elvira Jaller
Carlos Vargas
Hernán Carranza
Jorge Otero
Pilar Archila
Maritza Bermudez
Tatiana Gamez
Alessandro Russo
Umberto Malapelle
Diego de Miguel Perez
Vladmir C. Cordeiro de Lima
Helano Freitas
Erick Saldahna
Christian Rolfo
Rafael Rosell
CLICaP
Publikationsdatum
05.04.2023
Verlag
Springer International Publishing
Erschienen in
Targeted Oncology / Ausgabe 3/2023
Print ISSN: 1776-2596
Elektronische ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-023-00955-9

Weitere Artikel der Ausgabe 3/2023

Targeted Oncology 3/2023 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.