scholarly journals 1415TiP INSIGHT 2: Tepotinib + osimertinib in patients (pts) with EGFR-mutant NSCLC having acquired resistance to first-line osimertinib due to MET amplification (METamp)

2020 ◽  
Vol 31 ◽  
pp. S894
Author(s):  
E.F. Smit ◽  
E. Felip ◽  
N. Karachaliou ◽  
B. Ellers-Lenz ◽  
Y-L. Wu
2019 ◽  
Vol 12 (2) ◽  
pp. 625-630 ◽  
Author(s):  
Mike Ralki ◽  
Brigitte Maes ◽  
Karin Pat ◽  
Jokke Wynants ◽  
Kristof Cuppens

Epidermal growth factor receptor (EGFR)-targeted therapy has become standard of care in advanced stages EGFR-mutant non-small cell lung cancer. Acquired resistance to first-line EGFR-tyrosine kinase inhibitor (TKI) and subsequent disease progression is a common problem and mostly due to a secondary mutation (T790M) in EGFR. We report a case of a patient with EGFR-mutated lung adenocarcinoma who developed a complex resistance profile: T790M mutation, HER2 mutation and HER2 amplification after first-line EGFR-TKI. This patient was safely treated with a combination of osimertinib and trastuzumab and achieved a clinically meaningful and clear molecular response.This is the first reported case of acquired resistance to first-line EGFR-TKI based on three resistance mechanisms, treated with molecular targeted combination therapy.


2021 ◽  
Vol 16 (10) ◽  
pp. S1099-S1100
Author(s):  
C. Dooms ◽  
E. Nadal ◽  
J. Raskin ◽  
I. Demedts ◽  
J. Mazieres ◽  
...  

2019 ◽  
pp. 1-14 ◽  
Author(s):  
Sebastian Michels ◽  
Carina Heydt ◽  
Bianca van Veggel ◽  
Barbara Deschler-Baier ◽  
Nuria Pardo ◽  
...  

PURPOSE Third-generation epidermal growth factor receptor ( EGFR) tyrosine kinase inhibitors (TKIs) are effective in acquired resistance (AR) to early-generation EGFR TKIs in EGFR-mutant lung cancer. However, efficacy is marked by interindividual heterogeneity. We present the molecular profiles of pretreatment and post-treatment samples from patients treated with third-generation EGFR TKIs and their impact on treatment outcomes. METHODS Using the databases of two lung cancer networks and two lung cancer centers, we molecularly characterized 124 patients with EGFR p.T790M-positive AR to early-generation EGFR TKIs. In 56 patients, correlative analyses of third-generation EGFR TKI treatment outcomes and molecular characteristics were feasible. In addition, matched post-treatment biopsy samples were collected for 29 patients with progression to third-generation EGFR TKIs. RESULTS Co-occurring genetic aberrations were found in 74.4% of EGFR p.T790-positive samples (n = 124). Mutations in TP53 were the most frequent aberrations detected (44.5%; n = 53) and had no significant impact on third-generation EGFR TKI treatment. Mesenchymal-epithelial transition factor ( MET) amplifications were found in 5% of samples (n = 6) and reduced efficacy of third-generation EGFR TKIs significantly (eg, median progression-free survival, 1.0 months; 95% CI, 0.37 to 1.72 v 8.2 months; 95% CI, 1.69 to 14.77 months; P ≤ .001). Genetic changes in the 29 samples with AR to third-generation EGFR TKIs were found in EGFR (eg, p.T790M loss, acquisition of p.C797S or p.G724S) or in other genes (eg, MET amplification, KRAS mutations). CONCLUSION Additional genetic aberrations are frequent in EGFR-mutant lung cancer and may mediate innate and AR to third-generation EGFR TKIs. MET amplification was strongly associated with primary treatment failure and was a common mechanism of AR to third-generation EGFR TKIs. Thus, combining EGFR inhibitors with TKIs targeting common mechanisms of resistance may delay AR.


2020 ◽  
Author(s):  
Danlei Yu ◽  
Yiting Li ◽  
Kevin D-H Sun ◽  
Jiajia Gu ◽  
Zhen Chen ◽  
...  

Abstract Background The oncogenic protein, MET (or c-MET), is involved in the positive regulation of cell survival and proliferation and in mediating acquired resistance to EGFR-TKIs including AZD9291 (osimertinib). Thus, MET inhibition is a promising strategy for overcoming acquired EGFR-TKI resistance due to MET amplification. HQP8361 (MK8033) is a novel and selective MET kinase inhibitor that has completed a phase I clinical trial. The current study focuses on determining the activity and mechanism of action of HQP8361 as a single agent and in combination with AZD9291 against human NSCLC cells, particularly EGFR-mutant NSCLCs with acquired resistance to AZD9291. Methods Drug effects on cell growth in vitro were evaluated by measuring cell number alterations and colony formation and in vivo with mouse xenogtaft models, respectively. Apoptosis was assessed with annexin V/flow cytomentry and protein cleavage. Protein alterations were detected with Western blotting. Protein degradation was determined by comparing protein half-lives and inhibiting proteasome. Gene overexpression and knockout were achieved with lentiviral infection and CRISPR/Cas9, respectively. Significance of differences between two tested groups was analyzed with two-sided unpaired Student's t tests. Results The majority of human NSCLC cell lines tested, including those harboring EGFR-activating mutations with acquired resistance to AZD9291, had very low or undetectable levels of MET and p-MET and were insensitive to HQP8361. However, AZD9291-resistant (AR) cell lines derived from the EGFR-mutant HCC827 cell line possessed high levels of MET and p-MET and responded to HQP8361 single agent and particularly to the combination of HQP8361 and AZD9291. The HQP8361 and AZD9291 combination synergistically decreased the survival of these HCC827/AR cell lines with enhanced induction of apoptosis that involved alteration of Bim and Mcl-1 levels via modulating their degradation. Moreover, the combination also very effectively inhibited the growth of HCC827/AR xenografts in nude mice. Conclusions These preclinical findings support the potential of HQP8361 in the treatment of NSCLCs with MET amplification or highly activated MET protein and, when combined with AZD9291, in overcoming acquired resistance to EGFR-TKIs due to MET amplification.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 9043-9043
Author(s):  
Liu Li ◽  
Jingjing Qu ◽  
Jianfu Heng ◽  
Chunhua Zhou ◽  
Yi Xiong ◽  
...  

9043 Background: MET amplification is an important mechanism mediating acquired resistance to EGFR tyrosine kinase inhibitors (TKI). Until now, no consensus exists on the standard treatment strategy for this subset of patients due to the lack of clinical data from large cohort or controlled trials. In our clinical practice, three regimens were commonly administered to patients after MET amplification-mediated EGFR-TKI progression: EGFR-TKI and MET-TKI combination therapy, MET-TKI monotherapy, or chemotherapy. Our study aimed to compare the effectiveness of these three regimens. Methods: Seventy patients with EGFR-mutant advanced NSCLC who progressed from prior EGFR-TKI through the acquisition of MET amplification and received treatment between March 2015 and March 2020 were included in this study. Of them, 38 received EGFR-TKI plus crizotinib, 10 received crizotinib monotherapy, and 22 received platinum-based doublet chemotherapy. Somatic mutation profiling was performed on blood and tissue biopsy samples. Resistance mechanisms to the combination targeted therapy were also explored in 12 patients. Results: The objective response rate (ORR) and disease control rate (DCR) were 47.5% and 84.0% for EGFR-TKI+crizotinib group, 40.0% and 70.0% for crizotinib monotherapy group, and 18.2% and 50.0% for chemotherapy group, respectively. The EGFR-TKI+crizotinib group had significantly better ORR (P = 0.026) and DCR (P = 0.016) than the chemotherapy group but was not statistically different from the crizotinib monotherapy group (ORR, P = 0.73; DCR, P = 0.39). Progression-free survival (PFS) was significantly longer for the EGFR-TKI+crizotinib group than those who received crizotinib monotherapy (5.0 vs 2.3 months, P = 0.004) or chemotherapy (5.0 vs 2.9 months, P = 0.036), but overall survival was comparable (10.0 vs 4.1 vs 8.5 months, P = 0.088). TP53 mutation (58.5%) and EGFR amplifications (42.9%) were the two common concurrent mutations in the three cohorts. PFS was significantly longer for patients with either concurrent TP53 mutation (n = 17) (6.0 vs 2.3 vs 2.9 months, P = 0.009) or concurrent EGFR amplification (n = 13) (5.0 vs 1.2 vs 2.4 months, P = 0.016) who received EGFR-TKI+crizotinib. Potential molecular mechanisms of acquired resistance to EGFR-TKI+crizotinib therapy included EGFR T790M (n = 2), EGFR L718Q (n = 1), EGFR S645C (n = 1), MET D1228H (n = 1), BRAF V600E (n = 1), NRAS Q61H (n = 1), and amplifications in KRAS (n = 2), ERBB2 (n = 1), CDK4 (n = 1), and MYC (n = 2). Conclusions: Our study provides real-world clinical evidence, in the largest cohort to date, that simultaneous inhibition of EGFR and MET improves clinical outcomes of patients with EGFR-mutant NSCLC who acquired MET amplification from prior EGFR-TKI therapy, indicating that combinatorial regimen of EGFR-TKI and MET-TKI could be a more effective therapeutic strategy in this subset of patients.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e21580-e21580
Author(s):  
Mariacarmela Santarpia ◽  
Maria Rosaria Valerio ◽  
Nicolò Borsellino ◽  
Andrea Girlando ◽  
Gianfranco Mancuso ◽  
...  

e21580 Background: EGFR tyrosine kinase inhibitors (TKIs) represent the standard first-line therapy for advanced non-small cell lung cancer (NSCLC) patients with activating EGFR mutations. However, despite initial marked responses, tumors invariably develop acquired resistance to TKIs. Oligoprogression is commonly observed during treatment with oncogene-directed therapies, including EGFR TKIs, and refers to patients who experience disease progression only in limited sites as a result of heterogeneous mechanisms of resistance. The use of local ablative treatments for these resistant lesions may extend the duration of TKI therapy and potentially improve long-term disease control and survival. We e retrospectively analyzed the efficacy of EGFR TKI therapy continuation with high-dose hypofractionated radiation therapy (RT), in EGFR-mutant NSCLC patients with oligoprogressive disease. Methods: Patients with metastatic EGFR mutant NSCLC who developed oligoprogression during first-line treatment with gefitinib were included in this analysis. We evaluated progression-free survival 1 (PFS 1), defined as the time from initiation of TKI therapy until development of oligoprogression or death, and PFS 2, defined as time of focal progression until further progression of disease or death. Overall survival and safety were also assessed. Results: Thirty-six patients were included in the study. The median PFS 1 was 12.5 (4.0-23.2) months. High-dose hypofractionated RT consisted of intensity-modulated RT in 23 patients (64%) and stereotactic radiotherapy in 13 cases (36%). The median PFS 2 was 6.3 (2-12.5) months. Overall survival was 38.7 months (9.0-46.3). The treatment was well-tolerated and no patient had to discontinue TKI therapy because of adverse events during radiotherapy. Conclusions: Our therapeutic strategy, including high-dose hypofractionated RT in addition to TKI therapy, was feasible in the clinical setting and was associated with significant prolongation of disease control and improvement of survival outcomes, while being associated with manageable side effects. Our study further support the use of definitive therapeutic approaches in oligoprogressive disease, especially in oncogene-driven tumors. Molecular profiling of metastatic sites remains crucial to identify novel biomarkers, involved in the development of acquired resistance and oligoprogression, that may be useful to select patients for local treatments.


Sign in / Sign up

Export Citation Format

Share Document