46
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Molecular predictors of EGFR-mutant NSCLC transformation into LCNEC after frontline osimertinib: digging under the surface

      letter

      Read this article at

      ScienceOpenPublisherPMC
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          With the introduction of the third-generation tyrosine kinase inhibitor (TKI) osimertinib for the first-line treatment of epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC), understanding underlying mechanisms of resistance becomes crucial in order to optimize treatment strategy. Histological transformation towards both small-cell lung cancer (SCLC) and squamous cell carcinoma have been described as resistance patterns featuring peculiar molecular landscapes, potentially detectable at diagnosis and becoming clinically evident after clonal selection and disease evolution. 1 , 2 To date, only anecdotal reports have described the histological transformation to large-cell neuroendocrine carcinoma (LCNEC) after EGFR TKIs, but no data on molecular profiling have been reported. An 80-year-old Caucasian man, former light smoker, presented in January 2019 with persistent fever, despite antibiotic therapy. A total-body computed tomography (CT) scan and a fluorine-18 fluorodeoxyglucose positron emission tomography revealed a right upper lobe mass, with bilateral hilar and mediastinal lymph nodes, and a single bone lesion at the sternal body. In March 2019, biopsy of both the lung mass and the sternal lesion were diagnostic for lung adenocarcinoma (stage IVa; cT2aN3M1b), harboring an EGFR exon 19 deletion (E746_A750del). Next-generation sequencing (NGS) (FoundationOne CDx, Penzberg, Germany) confirmed the presence of an EGFR exon 19 deletion and revealed TP53 (tumor protein p53), RB1 (retinoblastoma), PTEN (phosphate and tensin homolog gene) and CHEK2 (checkpoint kinase) mutations, as well as NKX2-1 (NK2 homeobox 1 gene) and TERC (telomerase RNA component) amplification. From March 2019 to May 2020, he received first-line osimertinib 80 mg once daily, experiencing partial response (PR) as best response, with a progression-free survival of 15 months. In May 2020, the CT scan revealed bilateral enlargement of internal mammary lymph nodes; re-biopsy revealed metastasis of neuroendocrine carcinoma characterized by medium and large cells, supporting the diagnosis of LCNEC. NGS analysis of the new disease site showed the same genomic alterations detected at baseline, with the addition of AKT and BCL2L2 (Bcl-2-like 2) amplification. Osimertinib was interrupted and the patient was treated with carboplatin-etoposide, experiencing PR after 3 months (Figure 1). Figure 1 Disease evolution during treatment and clinical history. Upper panel: morphological, immunohistochemical and NGS features of diagnostic biopsy (1) and of biopsy taken at progression (2). (1) Bone metastasis of lung adenocarcinoma on hematoxylin–eosin (A), with positive immunolabelling for TTF1 (B) and napsin (C), and negative for synaptophysin (D). NGS analysis was performed on the lung biopsy specimen. (2) Lymph node metastasis of large cell neuroendocrine carcinoma on hematoxylin–eosin (A); immunostaining for synaptophysin (B) and Ki67 (C); loss of staining for Rb in the neoplastic cells with internal positive control cells (D). Lower panel: on the left, CT scan images show primary right upper lobe mass before and after 3 months of osimertinib, with evidence of partial response. On the right, a new CT scan reveals disease progression at bilateral internal mammary lymph nodes; radiological evaluation demonstrates PR after 3months of carboplatin-etoposide. Arrows indicate target lesions. BCL2L2, Bcl-2-like 2; CHEK2, checkpoint kinase; EGFR, epidermal growth factor receptor; NGS, next-generation sequencing; NKX2-1, NK2 homeobox 1 gene; PTEN, phosphate and tensin homolog gene; Rb, retinoblastoma protein; RB1, retinoblastoma gene; TERC, telomerase RNA component; TP53, tumor protein p53. To the best of our knowledge, this is the first report of transformation into LCNEC after first-line osimertinib. Tissue-based molecular profiling performed at baseline and at disease progression suggests that LCNEC transformation might be triggered, and potentially predicted, by molecular mechanisms similar to those observed during SCLC evolution, i.e. TP53 and RB1 inactivation. 3 In addition, NGS analysis revealed the presence of molecular alterations frequently detected in LCNEC, such as PTEN mutation and NKX2-1 amplification. 4 Recent data indicate that intrinsic heterogeneity and pre-existent subclones might predict the main mechanism(s) leading to osimertinib resistance at later timepoint. 5 In this specific case, we might speculate that a subclonal cell population harboring molecular biomarkers of neuroendocrine differentiation present at the baseline and not detectable upon conventional histological examination, underwent progressive clonal evolution under the selective pressure of EGFR TKI treatment, finally leading to a pathological transition towards high-grade LCNEC. Baseline molecular heterogeneity, particularly the presence of molecular stigmata of potential transformation into high-grade neuroendocrine carcinoma (TP53 and RB1 alterations), might support the upfront application of broad, NGS-based, genomic profiling and would constitute a potential biological rationale for treatments combining chemotherapy and EGFR TKIs in such cases. 5

          Related collections

          Most cited references5

          • Record: found
          • Abstract: found
          • Article: not found

          Clonal History and Genetic Predictors of Transformation Into Small-Cell Carcinomas From Lung Adenocarcinomas.

          Purpose Histologic transformation of EGFR mutant lung adenocarcinoma (LADC) into small-cell lung cancer (SCLC) has been described as one of the major resistant mechanisms for epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). However, the molecular pathogenesis is still unclear. Methods We investigated 21 patients with advanced EGFR-mutant LADCs that were transformed into EGFR TKI-resistant SCLCs. Among them, whole genome sequencing was applied for nine tumors acquired at various time points from four patients to reconstruct their clonal evolutionary history and to detect genetic predictors for small-cell transformation. The findings were validated by immunohistochemistry in 210 lung cancer tissues. Results We identified that EGFR TKI-resistant LADCs and SCLCs share a common clonal origin and undergo branched evolutionary trajectories. The clonal divergence of SCLC ancestors from the LADC cells occurred before the first EGFR TKI treatments, and the complete inactivation of both RB1 and TP53 were observed from the early LADC stages in sequenced tumors. We extended the findings by immunohistochemistry in the early-stage LADC tissues of 75 patients treated with EGFR TKIs; inactivation of both Rb and p53 was strikingly more frequent in the small-cell-transformed group than in the nontransformed group (82% v 3%; odds ratio, 131; 95% CI, 19.9 to 859). Among patients registered in a predefined cohort (n = 65), an EGFR mutant LADC that harbored completely inactivated Rb and p53 had a 43× greater risk of small-cell transformation (relative risk, 42.8; 95% CI, 5.88 to 311). Branch-specific mutational signature analysis revealed that apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC)-induced hypermutation was frequent in the branches toward small-cell transformation. Conclusion EGFR TKI-resistant SCLCs are branched out early from the LADC clones that harbor completely inactivated RB1 and TP53. The evaluation of RB1 and TP53 status in EGFR TKI-treated LADCs is informative in predicting small-cell transformation.
            • Record: found
            • Abstract: found
            • Article: not found

            Concurrent RB1 and TP53 alterations define a subset of EGFR-mutant lung cancers at risk for histologic transformation and inferior clinical outcomes

            EGFR -mutant lung cancers are clinically and genomically heterogeneous with concurrent RB1/TP53 alterations identifying a subset at increased risk for small cell transformation. The genomic alterations that induce lineage plasticity are unknown. Patients with EGFR/RB1/TP53 -mutant lung cancers, identified by NGS from 2014–2018, were compared to patients with untreated, metastatic EGFR -mutant lung cancers without both RB1 - and TP53 -alterations. Time to EGFR-tyrosine kinase inhibitor (EGFR-TKI) discontinuation (TTD), overall survival, SCLC transformation rate, and genomic alterations were evaluated. Patients with EGFR/RB1/TP53 -mutant lung cancers represented 5% (43/863) of EGFR -mutant lung cancers but were uniquely at risk for transformation (18%, 7/39), with no transformations in EGFR -mutant lung cancers without baseline TP53 and RB1 alterations. Irrespective of transformation, patients with EGFR/TP53/RB1 -mutant lung cancers had a shorter TTD than EGFR / TP53 and EGFR -mutant only cancers (9.5 vs 12.3 vs 36.6 months respectively, p = 2×10 −9 ). The triple-mutant population had a higher incidence of whole genome doubling (WGD) compared to NSCLC and SCLC at large (80% vs 34%, p < 5×10 −9 ; vs 51%, p < 0.002 respectively) and further enrichment in triple-mutant cancers with eventual small cell histology (7/7 pre-transformed plus 4/4 baseline SCLC vs 23/32 never transformed respectively, p = 0.05). AID/APOBEC mutation signature was also enriched in triple-mutant lung cancers that transformed (FDR = 0.03). EGFR/TP53/RB1 -mutant lung cancers are at unique risk of histologic transformation, with 25% presenting with de novo SCLC or eventual small cell transformation. Triple-mutant lung cancers are enriched in WGD and AID/APOBEC hypermutation which may represent early genomic determinants of lineage plasticity.
              • Record: found
              • Abstract: found
              • Article: not found

              Tumor Analyses Reveal Squamous Transformation and Off-Target Alterations As Early Resistance Mechanisms to First-line Osimertinib in EGFR-Mutant Lung Cancer

              Purpose: Patterns of resistance to first-line osimertinib are not well-established and have primarily been evaluated using plasma assays which cannot detect histologic transformation and have differential sensitivity for copy number changes and chromosomal rearrangements. Experimental Design: To characterize mechanisms of resistance to osimertinib, patients with metastatic EGFR -mutant lung cancers who received osimertinib at Memorial Sloan Kettering and had next-generation sequencing performed on tumor tissue before osimertinib initiation and after progression were identified. Results: Among 62 patients who met eligibility critieria, histologic transformation, primarily squamous transformation, was identified in 15% of first-line osimertinib cases and 14% of later-line cases. Nineteen percent (5/27) of patients treated with first-line osimertinib had off-target genetic resistance (2 MET amplification, 1 KRAS mutation, 1 RET fusion, and 1 BRAF fusion) whereas 4% (1/27) had an acquired EGFR mutation ( EGFR G724S). Patients with squamous transformation exhibited considerable genomic complexity; acquired PIK3CA mutation, chromosome 3q amplification and FGF amplification were all seen. Patients with transformation had shorter time on osimertinib and shorter survival compared to patients with on-target resistance. Initial EGFR sensitizing mutation, time on osimertinib treatment and line of therapy also influenced resistance mechanism that emerged. The compound mutation EGFR S768 + V769L and the mutation MET H1094Y were identified and validated as resistance mechanisms with potential treatment options. Conclusion: Histologic transformation and other off-target molecular alterations are frequent early emerging resistance mechanisms to osimertinib and are associated with poor clinical outcomes.

                Author and article information

                Contributors
                Journal
                ESMO Open
                ESMO Open
                ESMO Open
                Elsevier
                2059-7029
                16 January 2021
                February 2021
                16 January 2021
                : 6
                : 1
                : 100028
                Affiliations
                [1 ]Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
                [2 ]Section of Pathology, Department of Diagnostic and Public Health, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
                [3 ]Thoracic Surgery Department, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
                [4 ]Radiology Department, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
                [5 ]Section of Radiotherapy, Department of Surgery and Oncology, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
                [6 ]Pulmonary Unit, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
                Author notes
                [†]

                Equal contribution (these authors share last co-authorship).

                Article
                S2059-7029(20)32893-3 100028
                10.1016/j.esmoop.2020.100028
                7814107
                33465742
                c41c66e1-4d73-4070-b115-0bd86beaec4b
                © 2020 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                Categories
                Correspondence

                Comments

                Comment on this article

                Related Documents Log