Abstract
Introduction
Methods
Results
Conclusions
Keywords
Introduction
Patients and Methods
Population
Genomic Analysis
Results
Clinical Characteristics

Molecular Characteristics of Kinase Fusions in EGFR+ NSCLC

Case | Age/Sex | First CGP | Time Between Samples (d) | Second CGP | Interval Treatment History, Duration and Clinician Assessed Response | ||||
---|---|---|---|---|---|---|---|---|---|
EGFR Mutation | Associated Mutations | EGFR Mutation | RTK/BRAF Fusion | Associated Mutations | TMB | EGFR TKI(s) Pre-second CGP Biopsy | |||
Paired Tissue/Tissue | |||||||||
1 | 55/M | E746_A750del | EGFR amp, EGFR ex19del, MET amp, CDKN2A loss, TP53 Y205H, RAD51 M1fs8 | 1129 | E746_A750del | CCDC6-RET | AKT2 amp, CCND3 amp, CCNE1 amp, BCL2L2 amp, NFKBIA amp, NKX2-1 amp, CDKN2A/B loss, TP53 Y205H, CBL Y371N, RAD51 M1fs8, SPAT1 R1077H | 3.5 | Erlotinib 9 mo PR |
2 | 73/F | E746_A750del | AKT2 amp, CCND1 amp, AXL amp, CCNE1 amp, CDK6 amp, FGF19/3/4 amp, CDKN2A/B loss, TP53 K120fs26, PRMB1 K416fs3, SMAD4 Y133fs8 | 488 | E746_A750del | CCDC6-RET | AKT2 amp, CCND1 amp, AXL amp, CCNE1 amp, CDK6 amp, FGF19/3/4 amp, CDKN2A/B loss, PARK2 splice site, TP53 K120fs26, PRMB1 K416fs3, SMAD4 Y133fs8 | 6.1 | Erlotinib 10 mo PR |
3 | 46/M | E746_A750del+ T790M | CDKN2A/B loss, TP53 C176Y | 296 | E746_A750del | FGFR3-TACC3 | APC truncation ex15 and E1295fs93, CDKN2A/B loss, RB1 S618, TP53 C176Y | 2.6 | Erlotinib 8 mo PR, ASP8273 8 mo PR (apparent T790M dropout) |
4 | 61/F | L747_T751>P + T790M | CTNNB1 S37C, APC I1307K, CDKN2A/B loss | 216 | L747_T751>P | AGK-BRAF | CTNNB1 S37C, APC I1307K, CDKN2A/B loss | 7.0 | Erlotinib 12 mo PR, osimertinib 7 mo PR (apparent T790M dropout) |
5 | 64/F | E746_T751>L | PTEN G127R, TP53 E198, APC T1556fs3 | 544 | E746_T751>L | TPM-NTRK1 | PTEN G127R, TP53 E198, APC T1556fs3 | 6.1 | Erlotinib 16mos, PR |
Paired Tissue/Blood | |||||||||
6 | 66/F | L858R | EGFR E709K, NFKB1A amp, NKX2-1 amp, ZNF217 amp, TP53 V272L | 426 | L858R | FGFR3-TACC3 | CDKN2A p16 D108N p14 R122Q, TP53 V272L | NA | Afatinib + cetuximab 10 mo SD |
7 | 63/F | E746_A750del | MYC amp, NFKBIA amp, NKX2-1 amp, TP53 P190L | 483 | E746_A750del | EML4-ALK | BRAF V600E, NRAS Q61H, TP53 P190L | NA | Erlotinib 5 mo PR, osimertinib 5 mo with response in all but 3 progressing lesions |
8 | 51/F | E746_A750del | TP53 Y163C, RB1 loss | 631 | E746_A750del | FGFR3-TACC3 | TP53 Y163C | NA | Afatinib 18 mo PR |
9 | 46/F | L747_T751>P | TP53 L330R (ALK negative by FISH) | 503 | L747_T751>P | EML4-ALK | TP53 L330R | NA | Afatinib 16 mo PR |
10 | 67/F | L858R | CTNNB1 S37C, MDM2 amp, PIK3CB E1047K, FRS2 amp (ALK negative by FISH) | 894 | L858R | PLEKHA7-ALK | MDM2 amp | NA | Erlotinib 12 mo PR, afatinib 2 mo, osimertinib 12 mo PR (apparent T790M dropout) |
Paired Blood/Blood | |||||||||
11 | 61/M | L858R | TP53 R273C | 224 | L858R | EML4-ALK | TP53 R273C | NA | Erlotinib 5 mo (discontinued due to toxicity), afatinib 4mos SD |
12 | 74/M | E746_A750del+ T790M | TP53 V172F | 155 | E746_A750del | FGFR3-TACC3 | TP53 A86fs55, TP53 V172F | NA | Erlotinib 5 mo PR, osimertinib 4 mo PR (apparent T790M dropout) |

Case | Age/Sex | Blood/Tissue | CGP | Interval Treatment History, Duration, and Clinician Assessed Response | |||
---|---|---|---|---|---|---|---|
EGFR Mutation | RTK Fusion | TMB | Potential Coresistance Mutations | ||||
BRAF fusions | |||||||
13 | 63/F | Tissue | L858R | AGAP3-BRAF | 1.7 | — | Unknown |
14 | 68/F | Tissue | E746_A750del | AGK-BRAF | 3.5 | — | Unknown |
15 | 69/F | Tissue | L858R | ARMC10-BRAF | 9.6 | — | Unknown |
16 | 61/F | Tissue | L858R | SALL2-BRAF | 0.0 | — | Erlotinib 30 mo PR |
17 | 74/M | Tissue | E746_S752>V | DOCK4-BRAF | 3.5 | — | Erlotinib 24 mo PR |
18 | 57/F | Tissue | L858R | EPS515-BRAF | 2.4 | — | Erlotinib 9 mo PR |
19 | 69/M | Tissue | E746_A750 del | GHR-BRAF | 3.6 | — | Unknown |
20 | 63/F | Tissue | L858R | KIAA1549-BRAF | 4.3 | — | Unknown |
21 | 68/M | Tissue | L858R | TRIM24-BRAF | 2.6 | — | Unknown |
RET fusions | |||||||
22 | 69/M | Tissue | E746_A750>IP | CCDC6-RET | 0.0 | — | Erlotinib 4 y SD |
23 | 62/F | Tissue | L858R | NCOA4-RET | 4.3 | — | Afatinib 20 mo SD |
24 | 70/F | Tissue | L858R | TRIM24-RET | 2.6 | — | Unknown |
25 | 46/M | Blood | E746_A750del + T790M | TRIM24-RET | NA | — | Erlotinib PR, EGFR targeted therapy trial, osimertinib 8 mo PR, progression suspected on osimertinib, but was continuing on osimertinib for a total of almost 13 mo at time fusion+ sample was collected |
ALK fusions | |||||||
26 | 68/M | Tissue | L747_A755>SRD | STRN-ALK | 1.7 | — | Erlotinib 5 mo PR (T790M+ by outside testing), CO-1686 4 mo with liver metastasis shrinkage, osimertinib 6 mo PR (apparent T790M dropout) |
27 | 62/M | Blood | L747_P753>S | EML4-ALK | NA | — | Afatinib |
28 | 60/F | Blood | L858R | TFG-ALK | NA | EGFR T790M, C797S/G, L718Q | Osimertinib |
FGFR3 fusions | |||||||
29 | 66/F | Tissue | L747_A750>P | FGFR3-TACC3 | 12.2 | — | Erlotinib 11 mo PR |
30 | 77/M | Blood | L747_T751>P | FGFR3-TACC3 | NA | — | Unknown |
EGFR fusions | |||||||
31 | 66/F | Tissue | L858R | EGFR-FGFR1 | 7.0 | — | Erlotinib (T790M+ by outside testing), osimertinib (apparent T790M dropout) |
Case With a Novel PLEKHA7-ALK Fusion as Resistance Mechanism to Osimertinib, Responding to Osimertinib + Alectinib

Case With NCOA4-RET Fusion as a Resistance Mechanism to Afatinib, Responding to Afatinib + Cabozantibib
Discussion
- Shekine Y.
- Pavlick D.
- Klempner S.J.
- Shekine Y.
- Pavlick D.
- Klempner S.J.
Supplementary Data
- Supplemental Figure 1
Distribution of fusion genes in the setting of EGFR primary mutations.
- Supplemental Figure 2
The elevation and resolution of creatinine phosphokinase (CPK) after the addition of full dose alectinib 600 mg twice daily to osimertinib 80 mg once daily in case 10.
- Supplemental Tables 1 and 2
References
- Acquired EGFR C797S mutation mediates resistance to AZD9291 in non–small cell lung cancer harboring EGFR T790M.Nat Med. 2015; 21: 560-562
- Emergence of novel and dominant acquired EGFR solvent-front mutations at Gly796 (G796S/R) together with C797S/R and L792F/H mutations in one EGFR (L858R/T790M) NSCLC patient who progressed on osimertinib.Lung Cancer. 2017; 108: 228-231
- Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer.Cancer Disc. 2016; 6: 1118-1133
- Mutation of MET Y1230 as an Acquired Mechanism of Crizotinib Resistance in NSCLC with MET Exon 14 Skipping.J Thorac Oncol. 2017; 12: e89-e90
- Circulating tumor DNA identifies EGFR coamplification as a mechanism of resistance to crizotinib in a patient with advanced MET-amplified lung adenocarcinoma.J Thorac Oncol. 2017; 12: e155-e157
- Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers.Sci Transl Med. 2012; 4: 120ra17
- Emergence of RET rearrangement co-existing with activated EGFR mutation in EGFR-mutated NSCLC patients who had progressed on first- or second-generation EGFR TKI.Lung Cancer. 2015; 89: 357-359
- Emergence of FGFR3-TACC3 fusions as a potential by-pass resistance mechanism to EGFR tyrosine kinase inhibitors in EGFR mutated NSCLC patients.Lung Cancer. 2017; 111: 61-64
- MA 12.05 Genomic profiling of EGFR T790M mutated non-small cell lung cancer to evaluate the mechanisms of resistance to osimertinib.J Thorac Oncol. 2017; 12: S1848-S1849
- Metastatic EML4-ALK fusion detected by circulating DNA genotyping in an EGFR-mutated NSCLC patient and successful management by adding ALK inhibitors: a case report.BMC Cancer. 2015; 16: 62
- BRAF Fusion as a novel mechanism of acquired resistance to vemurafenib in BRAFV600E mutant melanoma.Clin Cancer Res. 2017; 23: 5631-5638
- Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing.Nat Biotechnol. 2013; 31: 1023-1031
- Analytic validation of a clinical circulating tumor DNA assay for patients with solid tumors.Ann Oncol. 27, 2016; ([Internet]) (Accessed September 29, 2017)
- Urine test for EGFR analysis in patients with non-small cell lung cancer.J Thorac Dis. 2017; 9: S1323-S1331
- Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance.Nat Rev Clin Oncol. 2017; 14: 735-748
- ROS1 fusions rarely overlap with other oncogenic drivers in non–small cell lung cancer.J Thorac Oncol. 2017; 12: 872-877
- ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non-small cell lung cancer.Clin Cancer Res. 2013; 19: 4273-4281
- Intratumoral heterogeneity of ALK-rearranged and ALK/EGFR coaltered lung adenocarcinoma.J Clin Oncol. 2015; 33: 3701-3709
- OA 09.02 osimertinib resistance mediated by loss of EGFR T790M is associated with early resistance and competing resistance mechanisms.J Thorac Oncol. 2017; 12: S1767-S1768
- Outcomes in patients with non–small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: a genomic study.Lancet Respir Med. 2018; 6: 107-116
- PLEKHA7 is an adherens junction protein with a tissue distribution and subcellular localization distinct from ZO-1 and E-cadherin.PLoS ONE. 2010; 5: e12207
- Tolerable and effective combination of full-dose crizotinib and osimertinib targeting MET amplification sequentially emerging after T790M positivity in EGFR-mutant non–small cell lung cancer.J Thorac Oncol. 2017; 12: e85-e88
- The distribution of BRAF gene fusions in solid tumors and response to targeted therapy.Int J Cancer. 2016; 138: 881-890
- Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients.Nat Med. 2017; 23: 703-713
- BRAF in lung cancers: analysis of 23,396 cases reveals recurrent BRAF mutations, fusions, kinase duplications, and concurrent alterations.JCO Precis Oncol. 2018; https://doi.org/10.1200/PO.17.00172
- Regulation and function of Spalt proteins during animal development.Int J Dev Biol. 2009; 53: 1385-1398
- CRAF gene fusions in pediatric low-grade gliomas define a distinct drug response based on dimerization profiles.Oncogene. 2017; 36: 6348-6358
- Impact of EML4-ALK Variant on resistance mechanisms and clinical outcomes in ALK-positive lung cancer.J Clin Oncol. 2018; 36: 1199-1206
- Acquired BRAF V600E mutation as resistant mechanism after treatment with third-generation EGFR tyrosine kinase inhibitor.J Thorac Oncol. 2017; 12: e181-e182
- FGFR3-TACC3 fusion in solid tumors: mini review.Oncotarget. 2016; 7: 55924-55938
- FGFR3-TACC3 fusion proteins act as naturally occurring drivers of tumor resistance by functionally substituting for EGFR/ERK signaling.Oncogene. 2017; 36: 471-481
- Targeting RET-driven cancers: lessons from evolving preclinical and clinical landscapes.Nat Rev Clin Oncol. 2018; 15: 1510167
- A next-generation TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion-positive solid tumors.Cancer Disc. 2017; 7: 963-972
- Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK inhibitor entrectinib: combined results from two phase I trials (ALKA-372-001 and STARTRK-1).Cancer Disc. 2017; 7: 400-409
- Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children.N Engl J Med. 2018; 378: 731-739
- Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors.Sci Transl Med. 2011; 3: 75ra26
- Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers.Clin Cancer Res. 2013; 19: 2240-2247
- Acquired resistance of EGFR-mutant lung cancer to a T790M-specific EGFR inhibitor: emergence of a third mutation (C797S) in the EGFR tyrosine kinase domain.JAMA Oncol. 2015; 1: 982-984
- Novel mutations on EGFR Leu792 potentially correlate to acquired resistance to osimertinib in advanced NSCLC.J Thorac Oncol. 2017; 12: e65-e68
- Cis-oriented solvent-front EGFR G796S mutation in tissue and ctDNA in a patient progressing on osimertinib: a case report and review of the literature.Lung Cancer. 2017; 8: 241-247
- High MET amplification level as a resistance mechanism to osimertinib (AZD9291) in a patient that symptomatically responded to crizotinib treatment post-osimertinib progression.Lung Cancer. 2016; 98: 59-61
Article info
Publication history
Footnotes
Drs. Schrock and Zhu contributed equally to this paper.
Disclosure: Drs. Schrock, Madison, Ross, Miller, and Ali are employees and have equity interest in Foundation Medicine, Inc. Dr. Zhu has received personal fees from Astra Zeneca, Roche/Genentech, Takeda, Biocept, and TP Therapeutics. Dr. Klempner has received personal fees from Foundation Medicine, Inc.; and has consulting/advisory roles with Lilly Oncology, Astellas, and Boston Biomedical. Dr. Ou has received personal fees from Foundation Medicine, Inc., Roche/Genentech, Takeda/ARIAD, Pfizer, and Astra Zeneca. The remaining authors declare no conflict of interest.
This work was partially presented at World Conference Lung Conference, Yokohama, Japan 2017: Ou et al. Abstract MA 12.03.
Identification
Copyright
User license
Creative Commons Attribution – NonCommercial – NoDerivs (CC BY-NC-ND 4.0) |
Permitted
For non-commercial purposes:
- Read, print & download
- Redistribute or republish the final article
- Text & data mine
- Translate the article (private use only, not for distribution)
- Reuse portions or extracts from the article in other works
Not Permitted
- Sell or re-use for commercial purposes
- Distribute translations or adaptations of the article
Elsevier's open access license policy