Purpose: Previous genomic studies have identified two mutually exclusive molecular subtypes of large-cell neuroendocrine carcinoma (LCNEC): the RB1 mutated (mostly comutated with TP53) and the RB1 wild-type groups. We assessed whether these subtypes have a predictive value on chemotherapy outcome.

Experimental Design: Clinical data and tumor specimens were retrospectively obtained from the Netherlands Cancer Registry and Pathology Registry. Panel-consensus pathology revision confirmed the diagnosis of LCNEC in 148 of 232 cases. Next-generation sequencing (NGS) for TP53, RB1, STK11, and KEAP1 genes, as well as IHC for RB1 and P16 was performed on 79 and 109 cases, respectively, and correlated with overall survival (OS) and progression-free survival (PFS), stratifying for non–small cell lung cancer type chemotherapy including platinum + gemcitabine or taxanes (NSCLC-GEM/TAX) and platinum-etoposide (SCLC-PE).

Results:RB1 mutation and protein loss were detected in 47% (n = 37) and 72% (n = 78) of the cases, respectively. Patients with RB1 wild-type LCNEC treated with NSCLC-GEM/TAX had a significantly longer OS [9.6; 95% confidence interval (CI), 7.7–11.6 months] than those treated with SCLC-PE [5.8 (5.5–6.1); P = 0.026]. Similar results were obtained for patients expressing RB1 in their tumors (P = 0.001). RB1 staining or P16 loss showed similar results. The same outcome for chemotherapy treatment was observed in LCNEC tumors harboring an RB1 mutation or lost RB1 protein.

Conclusions: Patients with LCNEC tumors that carry a wild-type RB1 gene or express the RB1 protein do better with NSCLC-GEM/TAX treatment than with SCLC-PE chemotherapy. However, no difference was observed for RB1 mutated or with lost protein expression. Clin Cancer Res; 24(1); 33–42. ©2017 AACR.

Translational Relevance

Large-cell neuroendocrine carcinoma (LCNEC) is a rare subtype of lung cancer for which the optimal treatment in advanced disease is debated (i.e., non-small cell lung cancer (NSCLC) versus small-cell lung cancer (SCLC) type chemotherapy regimen). In this study, which is the largest stage IV LCNEC patients cohort (n = 79) with information about chemotherapy treatment outcome, analyzed by next-generation sequencing, we tested in the two recently identified molecular subtypes of LCNEC (RB1 and TP53 mutated or STK11/KEAP1 and TP53 mutated) are valuable for treatment decision. Our results indicate that patients with LCNEC tumors that harbor a wild-type and/or express RB1 have superior overall survival when treated with NSCLC chemotherapy compared with SCLC chemotherapy. These data strengthen the relevance for molecular profiling in LCNEC, besides the oncogenic alterations already screened for in routine practice (e.g., EGFR).

Large-cell neuroendocrine carcinoma (LCNEC) is a high-grade neuroendocrine carcinoma with non–small cell cytologic features that accounts for 1% to 3% of all lung cancers (1, 2). Similar to small-cell lung cancer (SCLC), LCNEC is a disease with a poor prognosis (2, 3). The diagnosis of LCNEC requires assessing both morphology and neuroendocrine differentiation by IHC (4, 5). Previously, we and others have shown that separation of LCNEC from SCLC and pulmonary carcinoids can be difficult even on resection specimens (6–10). In the current WHO classification, some of the features used to classify a tumor as LCNEC overlap with those applied for SCLC, NSCLC, and carcinoids (8).

To improve the separation of LCNEC from carcinoids on a biopsy specimen, the proliferation marker Ki-67 with a cutoff >20% was proposed (10); however, the differential diagnosis between LCNEC and SCLC remains an issue for pathologists, due to crush artefacts, distorted cytologic features of SCLC on large tissue samples (11), tumor heterogeneity (11), and overlap in cell and nuclear size between LCNEC and SCLC (9). This is further worsened by the fact that, at diagnosis, both SCLC and LCNEC are often metastasized, and commonly only one biopsy specimen is available for diagnosis (12). The identification of diagnostic markers to allow separation of LCNEC from SCLC is therefore an unmet need.

Chemotherapy treatment for LCNEC is a subject of debate since it seems to be less chemosensitive than SCLC. In the American Society of Clinical Oncology (ASCO) guideline, either platinum–etoposide chemotherapy (SCLC-PE) treatment or the same regimen as for non–small cell nonsquamous cell carcinoma is advised for LCNEC (13), although SCLC-PE is considered as the most appropriate (13). Nevertheless, recent studies indicate that patients with LCNEC have a more favorable outcome when treated with platinum–gemcitabine or taxane chemotherapy (NSCLC-GEM/TAX) compared with SCLC-PE (14–16). The molecular characteristics that may explain these differences in the response to different chemotherapies remain unknown.

Several next-generation sequencing (NGS) studies have shown that LCNEC tumors can be further subdivided into two mutually exclusive groups based on their mutational patterns (17, 18): one harboring inactivation of TP53 and STK11 and/or KEAP1 genes, and the other one enriched for inactivation of TP53 and RB1, a hallmark of SCLC. It has been hypothesized that these LCNEC subtypes may require different chemotherapy treatment (17). In this study, we tested whether the described molecular LCNEC subtypes may have an impact on the chemotherapy response.

Regulations

The study protocol was approved by the medical ethical committee of the Maastricht University Medical Centre (METC azM/UM 14-4-043) and performed according to the Dutch “Federa, Human Tissue and Medical Research: Code of conduct for responsible use (2011)” regulations not requiring patient informed consent.

Patient and tumor selection

In this retrospective population-based study, all data were retrieved from the Netherlands Cancer Registry and Netherlands Pathology Registry (PALGA, the nationwide registry of pathology in the Netherlands; ref. 19) as previously described (12). Data managers from the cancer registry retrospectively updated (2015) clinical data of all first-line chemotherapy-treated stage IV LCNEC patients (n = 232, Fig. 1). Available data included clinical characteristics, TNM stage, overall survival (OS), and progression-free survival (PFS) from date of diagnosis until first evidence of progression, death or last day of follow-up, and chemotherapy details. All patients received platinum doublet (cisplatin or carboplatin) chemotherapy treatment, further divided into three groups: “NSCLC-GEM/TAX” including gemcitabine, docetaxel, or paclitaxel; “NSCLC-PEM” including pemetrexed; and “SCLC-PE” including etoposide. NSCLC-PEM chemotherapy was separated from the other NSCLC regimens because of previously reported resistance in (large cell) neuroendocrine carcinomas (14, 20–23).

Figure 1.

Selection of patients and tumor slides for panel-consensus review and molecular analyses. Abbreviations: N, number; NSCLC NED, non-small cell lung carcinoma with IHC neuroendocrine differentiation; NET NOS, neuroendocrine tumor not otherwise specified.

Figure 1.

Selection of patients and tumor slides for panel-consensus review and molecular analyses. Abbreviations: N, number; NSCLC NED, non-small cell lung carcinoma with IHC neuroendocrine differentiation; NET NOS, neuroendocrine tumor not otherwise specified.

Close modal

Panel consensus pathology revision

From all histologic specimens, the original hematoxylin and eosin (HE) and IHC slides were collected. Subsequently, three pathologists (R.J. van Suylen, E. Thunnissen, M. den Bakker), who were blinded for clinical outcome, systematically scored all cases at a multi-head microscope for WHO 2015 criteria. Proliferative activity was evaluated by estimation of MIB1 and mitotic counting (mitoses/2 mm2; ref. 8). The MIB1 (Ki-67) staining was scored (<25%, >25%) when available (10, 24). Either >10 mitosis/2 mm2, abundant tumor necrosis, or a Ki-67 staining of >25% of tumor cells was sufficient to score for high-grade tumor (19, 24). Diagnoses were considered as consensus when at least two pathologists agreed, further referred to as panel consensus. All panel consensus LCNEC tumors were included for NGS and IHC staining analysis when formalin-fixed paraffin embedded (FFPE) tissue block(s) were available (n = 109; Fig. 1).

DNA isolation

Tumor macrodissection was performed aiming at a tumor cell content of at least 20%. DNA was extracted from four to eight 10-μm slides using the Maxwell FFPE LEV Automated DNA Extraction Kit (Promega Corporation). DNA concentration was measured using the QuantiFluor dsDNA Dye System (Promega Corporation).

Amplicon design and target enrichment

One hundred and sixty-nine amplicons of 150 base pairs (bp) in size were designed using the Qiagen GeneRead DNAseq Custom V2 Builder tool reference CNGHS-02445X-169 (GRCh37) covering the following exons: TP53, RB1, STK11, and KEAP1. This custom GeneRead amplicon-based custom panel covered 100% of the coding region (i.e., exonic) of TP53, 95% of RB1, 81% of STK11, and 95% of KEAP1. A validated in-house protocol (IARC) was used to perform multiplex PCR with four separate primer pools. Per pool, 5 μL of DNA diluted to a maximum of 4 ng/μL (0.60–4.0) were dispensed and air-dried. Subsequently, 5 μL of the PCR mix were added (containing 2.5 μL primer, 1 μL PCR mix, 0.34 μL HotStar Taq, and 1.16 μL H20) and the DNA was amplified in a 384-well plate as following: 15 minutes at 95°C, and 25 cycles of 15 seconds at 95°C and 4 minutes at 60°C, and 10 minutes at 72°C. After amplification, the PCR products were pooled into a single reaction per sample.

Library preparation and next-generation sequencing

The amplified PCR products were purified using NucleoMag NGS Clean-up and Size Select beads (Macherey-Nagel). Purified PCR products were quantified by Qubit DNA high-sensitivity assay kit (Invitrogen Corporation). A minimum of 100 ng of purified PCR product was included for library preparation with the NEBNext Fast DNA Library Prep Set (New England BioLabs, USA) following an in-house validated protocol (IARC). End-repair was performed and ligated to specific adapters and in-house prepared individual barcodes (Eurofins MWG Operon). Bead purification was applied to clean libraries and amplification was performed. Equimolarly pooled libraries were loaded on a 2% agarose gel for electrophoresis (220 V, 40 minutes). Using the GeneClean Turbo kit (MP Biomedicals) DNA fragments of 110 to 220 bp were recovered from the pooled libraries. Library quality and quantity were assessed on the Agilent 2100 Bioanalyzer on-chip electrophoresis (Agilent Technologies). Sequencing was performed on the Ion Torrent Proton Sequencer (Life Technologies Corp.), aiming for a minimum coverage of 250×, using the Ion PI Hi-Q OT2 200 Kit and the Ion PI Hi-Q sequencing 200 Kit with the Ion PI chip V3 (Life Technologies Corp.), following manufacturer's instructions.

Technical duplicates and bioinformatical analysis

Technical duplicates were included for all samples and processed in identical 96- and 384-well plates to prevent PCR errors. Sequencing data were aligned to the hg19 (GRCh37) reference genome and BAM files were generated using the Torrent Suite Software (v4.4.2). For all amplicon positions, the read depth was calculated using SAMtools (25) and samples with a median coverage lower than 250× were excluded. Needlestack (https://github.com/IARCbioinfo/needlestack; ref. 26) was used to call variants with default parameters except for the base-quality and the mapping-quality thresholds (10 and 1, respectively). Annotation was performed with ANNOVAR (27) using the PopFreqAll (popfreq_all_20150413), COSMIC v77, SIFT and Polyphen (dbnsfp30a) databases (28, 29). We only considered those mutations identified by Needlestack in the two technical duplicates. In addition, we excluded the ones with an allelic fraction lower than 5%, a relative-variant strand bias (RVSB) higher than 0.85, or those already reported as germline in any of the ExAC, ESP or 1000G populations with a frequency larger than 0.001 (30–32). In addition, all mutations had to either be (i) reported in the COSMIC database, or (ii) damaging mutations (ii) damaging mutations (stop, indels and splice), or (iii), missense mutations classified as deleterious by SIFT or Polyphen databases (Supplementary Data File S1; NGS data).

RB1 and P16 IHC and scoring

The N-terminal and C-terminal regions of the RB1 protein were targeted with antibody 4H1 (1:100, Cell Signaling Technology) and 13A10 (1:100, Leica Biosystems). In addition, the protein P16 was targeted with antibody JC8 (1:400, Santa Cruz Biotechnology). Three-micron-thick FFPE slides were stained using a Dako Autostainer Link 48 system with the EnVision FLEX visualization Kit (DAKO, Agilent) according to the standard protocols. For 13A10 and JC8 high-pH antigen retrieval was used, and for 4H1 low-pH antigen retrieval was used. Tonsillar tissue (control for P16/RB1) and tumor stromal cells (internal control for RB1) were included as positive controls. H-scores were calculated as a total score of the percentage of tumor cells with staining intensity 1 (weak nuclear staining) ×1, intensity 2 (moderate nuclear staining) ×2, and intensity 3 (strong nuclear staining) ×3 with a maximum score of 300. H-scores were evaluated for all RB1 and P16 markers by E-J. M. Speel who was blinded for all clinical, histopathologic, and mutational data.

FISH

To detect homozygous deletions of the RB1 gene, FISH was performed with the ZytoLight SPEC RB1/13a12 Dual Color Probe (Zytovision). Three-micron-thick sections were cut from FFPE tumors without a detectable RB1 mutation but with loss of RB1 protein expression. FISH slides were deparaffinized, air dried at room temperature, pretreated with 0.2 mol/L HCL for 20 minutes at room temperature, followed by incubation in 1 mol/L NaSCN for 30 minutes at 80°C. Subsequently, sections were treated with pepsin from porcine stomach mucosa (Sigma-Aldrich; 0.5 mg/mL in 0.14 mol/L NaCl pH 2), postfixed in 1% formaldehyde in PBS for 10 minutes, and dehydrated in 70% to 100% ethanol. The ZytoLight SPEC RB1/13a12 Dual Color Probe was added under a coverslip (undiluted, 6–10 μL). Denaturation of probe and target DNA was carried out simultaneously for 5 min at 85°C before hybridization overnight at 37°C in a humid chamber (Thermobrite). After removing the coverslips, slides were stringently washed to remove unhybridized probe in 2× SSC/0.3% NP40 at pH 7.0 at 73°C for 2 minutes. Slides were dehydrated in an ascending ethanol series (70%–100%) and mounted in 0.2 μg/mL DAPI-Vectashield (Vector Laboratories). Probe signals were scored using a DM 5000 B fluorescence microscope (Leica) with specific filter sets for rhodamine-, fluorescein-, and DAPI.

Each tumor was screened for the presence/absence of RB1 (orange) and 13q12 (green) signals, and the predominant copy number per nucleus was noted (e.g., 1:1, 1:2, 2:2). Normal tonsillar tissue and internal normal tissue parts in the tumor tissue served as internal controls with a predominant 2:2 pattern. A homozygous deletion was considered when no RB1 (orange) signal was observed in the tumor tissue. All cases were evaluated by J. Derks and E.-J. M. Speel.

Statistical analysis

All analyses were performed using SPSS (version 22 for Windows, Inc.). The χ2 and Fisher exact test were used to compare categorical data; the Wilcoxon signed-rank test was used for continuous variables. OS and PFS were analyzed using two-sided log-rank test and survival curves were estimated using the Kaplan–Meier method. To evaluate the predictive role of RB1 mutation and IHC status, a Cox regression model was used including an interaction term for the marker and the chemotherapy treatment. Two-sided P values <0.05 were considered significant.

Pathology revision and molecular characterization by next-generation sequencing

A panel of three expert pathologists (R.J. van Suylen, E. Thunnissen, and M. den Bakker) reviewed the 232 clinically annotated and initially classified as stage IV LCNEC, available in the Netherlands Pathology Registry. In total, 148 of them were confirmed as panel consensus LCNEC tumors (Fig. 1; Supplementary Data File S1). The fact that tumors reclassified as carcinoids (n = 9) had a longer survival than those confirmed as LCNEC (P = 0.008) supports the value of the pathology revision (Supplementary Fig. S1A and S1B). Of the 148 confirmed LCNECs, 79 tumors passed the quality controls for NGS analyses and were targeted sequenced for the coding regions of TP53, RB1, KEAP1, and STK11 (Supplementary Data File S1).

We obtained a median coverage of 2,850× (range, 261–6,870) per sample. Mutations in TP53 were present in 85% of the cases (n = 67), RB1 in 47% (n = 37), KEAP1 in 18% (n = 14), and STK11 in 10% (n = 8); five samples were wild-type for the four genes analyzed (Fig. 2). RB1 was coaltered with TP53 in 34 of the 37 RB1-mutated tumors; RB1 mutations were mutually exclusive with STK11 P = 0.006 but not with KEAP1 mutations P = 0.71.

Figure 2.

Overview of genomic profiles of the LCNEC cases analyzed by targeted exon sequencing of the RB1, TP53, STK11, KEAP1 genes and by IHC for RB1 and P16. In total, 79 LCNEC tumors were sequenced, and an additional set of 30 were only analyzed by IHC. *, In tumor sample 162 the DNA was isolated from tissue obtained 10 months after initiation of treatment.

Figure 2.

Overview of genomic profiles of the LCNEC cases analyzed by targeted exon sequencing of the RB1, TP53, STK11, KEAP1 genes and by IHC for RB1 and P16. In total, 79 LCNEC tumors were sequenced, and an additional set of 30 were only analyzed by IHC. *, In tumor sample 162 the DNA was isolated from tissue obtained 10 months after initiation of treatment.

Close modal

Clinical relevance of the mutational patterns of LCNEC tumors

The clinical characteristics of the patients, which tumors were sequenced, are shown in Table 1: median age was 64 (range, 51–79), 65% were males, 55% completed first-line chemotherapy (≥4 cycles), and 19% received second-line chemotherapy treatment. When considering only the patients with available data on the subtype of chemotherapy (n = 72, 91%), we observed that those LCNEC tumors that harbored a wild-type RB1 gene showed a significant longer OS when treated with NSCLC-GEM/TAX compared with SCLC-PE [(9.6; 95% confidence interval (CI), 7.7–11.6 versus 5.8 (95% CI, 5.5–6.1) months, P = 0.026] and to NSCLC-PEM chemotherapy [6.7 (95% CI, 5.1–8.2), P = 0.039; Fig. 3A)]. No difference was observed in the case of LCNECs with an RB1 mutation (Fig. 3B). Using a Cox regression model, the presence of a wild-type RB1 was associated with a significant difference (HR, 2.37; 95% CI, 1.09–5.19) favoring NSCLC-GEM/TAX chemotherapy over SCLC-PE treatment. However, comparison of the overall group did not identify a significant interaction between the RB1 mutational status and the chemotherapy treatment (P = 0.35; Fig. 4).

Table 1.

Clinical characteristics of patients included for NGS and IHC analyses

NGSPIHCP
Clinical characteristicsTotalRB1wtRB1mtwt vs. mtTotalRB1+RB1+ vs.
Total patients included, n 79 42 37  109 31 78  
Age (median, IQR)a 65 (51–79) 64 (46–82) 65 (53–77) 0.87 64 (59–79) 64 (52–76) 63 (48–78) 0.99 
Gender    0.37    0.33 
 Male 51 (65) 29 (69) 22 (59)  66 (61) 21 (68) 45 (58)  
 Female 28 (35) 13 (31) 15 (41)  43 (39) 10 (32) 33 (42)  
Chemotherapy clusters    0.91b    0.56b 
 NSCLC-GEM/TAX 31 (39) 15 (35) 16 (43)  45 (41) 14 (45) 31 (40)  
 SCLC-CE 28 (35) 13 (31) 15 (41)  40 (37) 9 (29) 31 (40)  
 NSCLC-PEM 13 (17) 7 (17) 6 (16)  15 (14) 3 (10) 12 (15)  
 Unknown 7 (9) 7 (17) 0 (0)  9 (8) 5 (16) 4 (5)  
Chemotherapy subtypes    —    – 
 Gemcitabine 22 (28) 11 (26) 11 (30)  35 (32) 9 (29) 26 (35)  
 Taxanes (docetaxel/paclitaxel) 9 (11) 4 (9) 5 (14)  10 (9) 5 (16) 5 (7)  
 Etoposide 28 (35) 13 (30) 15 (40)  40 (37) 9 (29) 31 (41)  
 Pemetrexed 13 (16) 7 (17) 6 (16)  15 (14) 3 (10) 12 (16)  
 Unknown 7 (9) 7 (17) 0 (0)  9 (8) 5 (16) 1 (1)  
Cycles of chemotherapy    0.47c    0.56c 
 1 9 (11) 5 (12) 4 (11)  17 (16) 5 (16) 12 (15)  
 2 13 (17) 5 (12) 8 (22)  13 (12) 2 (7) 11 (14)  
 3 12 (15) 5 (12) 7 (19)  14 (13) 5 (16) 9 (12)  
 4 34 (43) 20 (47) 14 (37)  49 (45) 13 (42) 36 (46)  
>4 9 (11) 5 (12) 4 (11)  13 (12) 4 (13) 9 (12)  
 Unknown 2 (3) 2 (5) 0 (0)  3 (3) 2 (6) 1 (1)  
Second-line chemotherapy treatment    0.99    0.21 
 No 64 (81) 34 (81) 30 (81)  89 (82) 23 (74) 66 (85)  
 Yes 15 (19) 8 (19) 7 (19)  20 (18) 8 (26) 12 (15)  
NGSPIHCP
Clinical characteristicsTotalRB1wtRB1mtwt vs. mtTotalRB1+RB1+ vs.
Total patients included, n 79 42 37  109 31 78  
Age (median, IQR)a 65 (51–79) 64 (46–82) 65 (53–77) 0.87 64 (59–79) 64 (52–76) 63 (48–78) 0.99 
Gender    0.37    0.33 
 Male 51 (65) 29 (69) 22 (59)  66 (61) 21 (68) 45 (58)  
 Female 28 (35) 13 (31) 15 (41)  43 (39) 10 (32) 33 (42)  
Chemotherapy clusters    0.91b    0.56b 
 NSCLC-GEM/TAX 31 (39) 15 (35) 16 (43)  45 (41) 14 (45) 31 (40)  
 SCLC-CE 28 (35) 13 (31) 15 (41)  40 (37) 9 (29) 31 (40)  
 NSCLC-PEM 13 (17) 7 (17) 6 (16)  15 (14) 3 (10) 12 (15)  
 Unknown 7 (9) 7 (17) 0 (0)  9 (8) 5 (16) 4 (5)  
Chemotherapy subtypes    —    – 
 Gemcitabine 22 (28) 11 (26) 11 (30)  35 (32) 9 (29) 26 (35)  
 Taxanes (docetaxel/paclitaxel) 9 (11) 4 (9) 5 (14)  10 (9) 5 (16) 5 (7)  
 Etoposide 28 (35) 13 (30) 15 (40)  40 (37) 9 (29) 31 (41)  
 Pemetrexed 13 (16) 7 (17) 6 (16)  15 (14) 3 (10) 12 (16)  
 Unknown 7 (9) 7 (17) 0 (0)  9 (8) 5 (16) 1 (1)  
Cycles of chemotherapy    0.47c    0.56c 
 1 9 (11) 5 (12) 4 (11)  17 (16) 5 (16) 12 (15)  
 2 13 (17) 5 (12) 8 (22)  13 (12) 2 (7) 11 (14)  
 3 12 (15) 5 (12) 7 (19)  14 (13) 5 (16) 9 (12)  
 4 34 (43) 20 (47) 14 (37)  49 (45) 13 (42) 36 (46)  
>4 9 (11) 5 (12) 4 (11)  13 (12) 4 (13) 9 (12)  
 Unknown 2 (3) 2 (5) 0 (0)  3 (3) 2 (6) 1 (1)  
Second-line chemotherapy treatment    0.99    0.21 
 No 64 (81) 34 (81) 30 (81)  89 (82) 23 (74) 66 (85)  
 Yes 15 (19) 8 (19) 7 (19)  20 (18) 8 (26) 12 (15)  

Abbreviations: IQR, interquartile range; wt, wild-type; mt, mutation.

aWilcoxon signed-rank test.

bExcluded unknown cases for comparison.

cExcluded unknown cases for comparison, compared ≤2 vs. >2 cycles.

Figure 3.

Overall survival for subtypes of chemotherapy in panel consensus LCNEC with RB1 wild-type* (A), RB1 mutation (B), H-score ≥50 for RB1* IHC (C), H-score <50 for RB1 IHC (D), H-score <50 for P16 IHC (E), and H-score ≥50 for RB1* or <50 for P16 on IHC analysis (F). Abbreviations: No, number of. *, case 162 excluded from analyses.

Figure 3.

Overall survival for subtypes of chemotherapy in panel consensus LCNEC with RB1 wild-type* (A), RB1 mutation (B), H-score ≥50 for RB1* IHC (C), H-score <50 for RB1 IHC (D), H-score <50 for P16 IHC (E), and H-score ≥50 for RB1* or <50 for P16 on IHC analysis (F). Abbreviations: No, number of. *, case 162 excluded from analyses.

Close modal
Figure 4.

Cox regression model for overall survival including the covariates RB1 and chemotherapy. A test for interaction was performed to evaluate the predictive value of RB1 mutations and RB1 protein expression measured by IHC, for chemotherapy outcome.

Figure 4.

Cox regression model for overall survival including the covariates RB1 and chemotherapy. A test for interaction was performed to evaluate the predictive value of RB1 mutations and RB1 protein expression measured by IHC, for chemotherapy outcome.

Close modal

The PFS of RB1 wild-type NSCLC-GEM/TAX–treated patients was also significantly higher compared with treatment with SCLC-PE [6.1 (95% CI, 4.2–8.0) months versus 5.7 (95% CI, 3.9–7.6), P = 0.019] but similar to treatment with NSCLC-PEM [4.7 (95% CI, 3.0–6.4), P = 0.18; Supplementary Fig. S2A]. Similarly to OS, no difference was observed for PFS in RB1-mutated LCNEC patients for the different chemotherapy regimens (Supplementary Fig. S2B). Finally, the mutational status of none the assessed genes (TP53, RB1, STK11, KEAP1) had a prognostic value (Fig. 5A–D).

Figure 5.

A–D, Overall survival for mutational status in panel consensus LCNEC (N = 78)*. E and F, Overall survival in panel consensus LCNEC for RB1* (N = 108) and P16* (N = 107) IHC H-score. Abbreviations: No, number of. *, case 162 excluded for analyses.

Figure 5.

A–D, Overall survival for mutational status in panel consensus LCNEC (N = 78)*. E and F, Overall survival in panel consensus LCNEC for RB1* (N = 108) and P16* (N = 107) IHC H-score. Abbreviations: No, number of. *, case 162 excluded for analyses.

Close modal

Correlation between mutational patterns and IHC analyses

We assessed the RB1 protein expression levels by IHC in all of the 79 tumors sequenced (Fig. 2) and found that 92% of the RB1-mutated LCNEC tumors had an RB1 H-score of 0 [the median was 0 (range, 0–200) compared with the internal controls; Supplementary Fig. S3A]. The three cases that retained RB1 expression carried two splice mutations and one single nucleotide variation, all reported in COSMIC, respectively (Supplementary Data File S1). No difference was observed regarding the C- and N-terminal RB1 protein staining when stratifying by type of mutation (splice/indel/single nucleotide variants).

In LCNEC tumors with RB1 wild-type, the median H-score was 50 (range, 0–200; Fig. 2; Supplementary Data File S1, Supplementary Fig. S3C). In 18 (43%), the H-score was 0. To test whether a copy-number alteration was the underlying mechanism for this loss of expression, we performed FISH in the 16 cases for which tissue was available (Supplementary Data File S1). However, no homozygous deletion was found in any of the samples suggesting alternate mechanisms for RB1 inactivation, such as genomic rearrangements or large deletions detectable with the techniques used in this study.

Considering the reported interobserver variation in the diagnosis of SCLC and LCNEC, and the fact that virtually all SCLCs carry an inactivated RB1 (33), we evaluated whether there was a correlation between the panel diagnosis and the loss of RB1 protein expression. In total, we performed IHC analyses on 109 panel consensus diagnosed LCNEC. For 98 of them, the panel unanimously diagnosed the tumors as LCNEC but for 11, one of the pathologists classified the tumor as SCLC. We did not observe a significant difference in the prevalence of RB1 expression in these 11 cases (27% with an H-score >50) when comparing with the 98 LCNEC unanimously classified (29% with an H-score >50, P = 0.93).

Previous studies have identified a correlation between RB1 inactivation, loss of expression, and, P16 expression (34, 35). We, therefore, analyzed our series of samples for P16 protein expression and found that, while the median P16 H-score in the RB1 wild-type LCNEC group was 180 (range, 0–300), this value went up to 300 (range, 0–300) in the RB1-mutated cases (Fig. 2; Supplementary Fig. S3B). In total, 91% of the LCNEC tumors with a P16 H-score <50 harbored a wild-type RB1 gene (Fig. 2; Supplementary Fig. S3D).

Clinical relevance of the IHC results

Patients with LCNEC showing an RB1 H-score ≥50 had a significant longer OS when treated with NSCLC-GEM/TAX than with SCLC-PE [9.6 (95% CI, 7.4–11.8) vs. 1.9 (95% CI, 1.7–2.1) months, P = 0.001] or NSCLC-PEM [4.8 (95% CI, 3.9–5.7) months, P = 0.007; Fig. 3C]. Cox regression analysis confirmed the predictive value of RB1-positive staining on NSCLC-GEM/TAX versus SCLC-PE chemotherapy outcome (HR 4.96; 95% CI, 1.79–13.74, Pinteraction = 0.002; Fig. 4). PFS was also significantly longer for NSCLC-GEM/TAX versus SCLC-PE–treated patients [5.5 (95% CI, 1.9–9.0) and 1.7 (95% CI, 0.0–4.8) months (P = 0.023), respectively] but not versus NSCLC-PEM [4.1 (95% CI, 4.0–4.2) months, P = 0.21; Supplementary Fig. S2C]. No statistically significant difference in response to different chemotherapy treatments was observed in patients with LCNEC tumors with an RB1 H-score <50, independently of their mutational status (Fig. 3D; Supplementary Fig. S2D).

P16 IHC H-score <50 was correlated with improved OS for NSCLC-GEM-TAX versus SCLC-PE chemotherapy (P = 0.028; Fig. 3D) but it had no impact on PFS (P = 0.24; Supplementary Fig. S2D). Combined evaluation of RB1 H-score ≥50 and/or P16 <50 showed identical results for OS (P = 0.002; Fig. 3F) and PFS (P = 0.027; Supplementary Fig. S2F). Similarly to the mutational status, none of the RB1 and P16 H-scores had prognostic value (Fig. 5E and F).

Once diagnosed, LCNEC is frequently treated with SCLC-PE chemotherapy, with poor responses (13, 14, 36, 37). Recently, we provided evidence that NSCLC-GEM/TAX chemotherapy may perform better than SCLC-PE chemotherapy on LCNEC tumors (14). Here we have tested whether the molecular characteristics of the LCNEC tumors might explain these differences.

In line with what has been already reported (17, 18, 38), TP53 was found mutated in 85% of our LCNEC cases, RB1 in 47%, KEAP1 in 18%, and STK11 in 10%. RB1 was coaltered with TP53 in 92% of the RB1-mutated tumors, and mutations in RB1 and STK11 occurred in a mutually exclusive way. The frequency of STK11 mutations was lower than expected, probably due to the fact that with our approach we could only cover 60% of the coding region of this gene. Although the mutational status of the genes analyzed did not have a prognostic value, we observed that patients with RB1 wild-type LCNECs showed a significant longer OS when treated with NSCLC-GEM/TAX compared with SCLC-PE and to NSCLC-PEM chemotherapies.

RB1 is inactivated in virtually all SCLCs but only in half of LCNECs (17, 33, 38–40). Similarly, RB1 protein expression is more frequently lost in SCLC (∼90%) than in LCNEC (45%–67%; refs. 17, 18, 35, 38, 41). However, its clinical relevance has not been assessed thoroughly but for a recent prospective study on SCLC reporting that patients with wild-type RB1 but loss of the protein expression showed inferior OS and PFS when treated with SCLC-PE chemotherapy (42). In our study, we found that RB1 expression was completely lost in almost all RB1-mutated LCNECs, but also in 47% of the wild-type cases. Homozygous gene deletions measured by FISH did not explain the loss of expression in the wild-type samples. This suggests that protein expression might be a more reliable measurement of RB1 inactivation than mutation. In total, RB1 protein expression was strongly downregulated or completely lost in 72% of the LCNEC tumors analyzed, similarly to what has been reported in recent studies (17, 38). Similarly to RB1 wild-type LCNEC patients, those with RB1-expressing tumors also showed a significant longer OS when treated with NSCLC-GEM/TAX than with SCLC-PE or NSCLC-PEM. But, in addition, Cox regression analysis confirmed the predictive value of RB1-positive staining on NSCLC-GEM/TAX versus SCLC-PE chemotherapy outcome, and PFS was also significantly longer for NSCLC-GEM/TAX versus SCLC-PE–treated patients.

P16 (CDKN2A) functions as an inhibitor of cyclin D-dependent kinases (CDK4/6) that phosphorylates RB1 enabling cell proliferation (43). CDKN2A (P16) and RB1 inactivation seem to be mutually exclusive in LCNEC (18, 44), and their expression is strongly correlated; previous studies have indicated that combined low RB1 and high P16 proteins expression is observed in 45% to 78% of LCNEC and almost always in SCLC (>90%; refs. 35, 38, 41, 43). In our data, we have validated this patter, and we have additionally found that combined RB1 expression and loss of P16 expression are predictive for improved outcome on NSCLC-GEM/TAX chemotherapy treatment, although P16 staining did not show additive value to the RB1 staining alone.

Overall, we found that RB1 mutational status and RB1/P16 protein expression are predictive markers for chemotherapy response and may aid to guide therapeutic decisions in advanced LCNEC disease. This is the largest (population-based) study evaluating chemotherapy outcome related to mutational patterns in panel-reviewed LCNECs. Although these results are of great interest for the clinical management of LCNEC, the few limitations of our study due to its retrospective design encourage the replication of these results in a prospective randomized clinical trial that stratifies LCNEC based on genomic subtypes and by RB1/P16 protein expression, and investigate outcome to NSCLC-GEM/TAX and SCLC-PE chemotherapy subtypes. In addition, these markers could be tested in biopsy specimens of high-grade neuroendocrine carcinomas to evaluate whether they could help in the differential diagnosis of LCNEC and SCLC.

E.M. Speel reports receiving other commercial research support from Bristol-Myers Squibb. No potential conflicts of interest were disclosed by the other authors.

Conception and design: J.L. Derks, E. Thunnissen, H.J.M. Groen, E.F. Smit, L. Fernandez-Cuesta, E.-J.M. Speel, A.-M.C. Dingemans

Development of methodology: J.L. Derks, E. Thunnissen, M. Foll, J.D. McKay, L. Fernandez-Cuesta, E.-J.M. Speel, A.-M.C. Dingemans

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.L. Derks, N. Leblay, E. Thunnissen, M. den Bakker, R. Damhuis, E.C. van den Broek, A. Chabrier, J.D. McKay, E.-J.M. Speel, A.-M.C. Dingemans

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.L. Derks, N. Leblay, E. Thunnissen, R.J. van Suylen, H.J.M. Groen, R. Damhuis, M. Foll, L. Fernandez-Cuesta, E.-J.M. Speel, A.-M.C. Dingemans

Writing, review, and/or revision of the manuscript: J.L. Derks, E. Thunnissen, R.J. van Suylen, M. den Bakker, H.J.M. Groen, E.F. Smit, R. Damhuis, E.C. van den Broek, A. Chabrier, M. Foll, L. Fernandez-Cuesta, E.-J.M. Speel, A.-M.C. Dingemans

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): J.L. Derks, E. Thunnissen, R.J. van Suylen, R. Damhuis, E.C. van den Broek, A. Chabrier, A.-M.C. Dingemans

Study supervision: L. Fernandez-Cuesta, E.-J.M. Speel, A.-M.C. Dingemans

Other (specimen review): M. den Bakker

We would like to thank G. Roemen and A. Haesevoets of the Department of Pathology of the Maastricht University Medical Centre (Maastricht, the Netherlands) for their help with performing the FISH analyses.

PALGA-group co-authors: L. Arensman, Meander Medisch Centrum Klinische Pathologie; F.E. Bellot, Klinische Pathologie Hoofddorp; J.E. Broers, Isala Klinieken; C.M. van Dish; Klinische pathologie Groene Hart Ziekenhuis; K.E.S.Duthoi, Amphia Ziekenhuis; M.J. Flens, Symbiant; J.M.M. Grefte, Gelre Ziekenhuis Klinische Pathologie; M.C.H. Hogenes, Labpon; R. Natté, Haga Ziekenhuis; A.F. van Hamel, Pathologie SSZOG; P.J.J.M. Klinkhamer, Stichting PAMM; J.W.R. Meijer, Ziekenhijs Rijnstate; J.C.C.van der Meij, Pathologie Friesland; F.H. van Nederveen Laboratorium voor Pathologie (PAL); E.W.P. Nijhuis, Onze Lieve Vrouwe Gasthuis; M.F.M. van Oosterhout, St. Antonius Ziekenhuis; S.H. Sastrowijoto, Pathologie Sittard; K. Schelfout, Stichting Pathologisch en Cytologisch Laboratorium West-Brabant; J. Sietsma, Pathologie Martini Ziekenhuis; F.M.M. Smedts, Reinier Haga MDC; M.M. Smits, Laboratorium voor Klinische Pathologie; J. Stavast, Laboratorium Klinische Pathologie Centraal Brabant; W. Timens, Pathologie UMCG; M.L. van Velthuysen, NKI-AVL; A. Vink, Universitair Medisch Centrum Utrecht; C.C.A.P. Wauters, Canisius Wilhelmina Ziekenhuis; S. Wouda, VieCuri Medical Centre.

This study was supported by grants from the Dutch Cancer Society (UM-2014-7110, to A-M. C. Dingemans). J.L. Derks is the recipient of an ERS/EMBO Joint Research Fellowship (STRTF 2016) 7178. The research leading to these results has received funding from the European Respiratory Society (ERS) and European Molecular Biology Organization (EMBO)”.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Takei
H
,
Asamura
H
,
Maeshima
A
,
Suzuki
K
,
Kondo
H
,
Niki
T
, et al
Large cell neuroendocrine carcinoma of the lung: a clinicopathologic study of eighty-seven cases
.
J Thorac Cardiovasc Surg
2002
;
124
:
285
92
.
2.
Derks
JL
,
Hendriks
LE
,
Buikhuisen
WA
,
Groen
HJ
,
Thunnissen
E
,
van Suylen
RJ
, et al
Clinical features of large cell neuroendocrine carcinoma: a population-based overview
.
Eur Respir J
2016
;
47
:
615
24
.
3.
Asamura
H
,
Kameya
T
,
Matsuno
Y
,
Noguchi
M
,
Tada
H
,
Ishikawa
Y
, et al
Neuroendocrine neoplasms of the lung: a prognostic spectrum
.
J Clin Oncol
2006
;
24
:
70
6
.
4.
Travis
WD
,
Brambilla
E
,
Burke
AP
,
Marx
A
,
Nicholson
AG
.
WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart
. Fourth edition. In:
WHO Classification of Tumours
.
Geneva, Switzerland
:
World Health Organization
; 
2015
.
5.
Travis
WD
,
Linnoila
RI
,
Tsokos
MG
,
Hitchcock
CL
,
Cutler
GB
 Jr
,
Nieman
L
, et al
Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases
.
Am J Surg Pathol
1991
;
15
:
529
53
.
6.
Travis
WD
,
Gal
AA
,
Colby
TV
,
Klimstra
DS
,
Falk
R
,
Koss
MN
. 
Reproducibility of neuroendocrine lung tumor classification
.
Hum Pathol
1998
;
29
:
272
9
.
7.
Den Bakker
MA
,
Willemsen
S
,
Grunberg
K
,
Noorduijn
LA
,
Van Oosterhout
MFM
,
Van Suylen
RJ
, et al
Small cell carcinoma of the lung and large cell neuroendocrine carcinoma interobserver variability
.
Histopathology
2010
;
56
:
356
63
.
8.
Thunnissen
E
,
Borczuk
AC
,
Flieder
DB
,
Witte
B
,
Beasley
MB
,
Chung
JH
, et al
The use of immunohistochemistry improves the diagnosis of small cell lung cancer and its differential diagnosis. An international reproducibility study in a demanding set of cases
.
J Thorac Oncol
2017
;
12
:
334
46
.
9.
Marchevsky
AM
,
Gal
AA
,
Shah
S
,
Koss
MN
. 
Morphometry confirms the presence of considerable nuclear size overlap between "small cells" and "large cells" in high-grade pulmonary neuroendocrine neoplasms
.
Am J Clin Pathol
2001
;
116
:
466
72
.
10.
Fabbri
A
,
Cossa
M
,
Sonzogni
A
,
Papotti
M
,
Righi
L
,
Gatti
G
, et al
Ki-67 labeling index of neuroendocrine tumors of the lung has a high level of correspondence between biopsy samples and surgical specimens when strict counting guidelines are applied
.
Virchows Arch
2017
;
470
:
153
64
.
11.
Nicholson
SA
,
Beasley
MB
,
Brambilla
E
,
Hasleton
PS
,
Colby
TV
,
Sheppard
MN
, et al
Small cell lung carcinoma (SCLC): a clinicopathologic study of 100 cases with surgical specimens
.
Am J Surg Pathol
2002
;
26
:
1184
97
.
12.
Derks
JL
,
Jan van Suylen
R
,
Thunnissen
E
,
den Bakker
MA
,
Smit
EF
,
Groen
HJ
, et al
A population-based analysis of application of WHO nomenclature in pathology reports of pulmonary neuroendocrine tumors
.
J Thorac Oncol
2016
;
11
:
593
602
.
13.
Masters
GA
,
Temin
S
,
Azzoli
CG
,
Giaccone
G
,
Baker
S
 Jr
,
Brahmer
JR
, et al
Systemic therapy for stage IV non-small-cell lung cancer: American Society of Clinical Oncology Clinical Practice Guideline Update
.
J Clin Oncol
2015
;
33
:
3488
515
.
14.
Derks
JL
,
van Suylen
RJ
,
Thunnissen
E
,
den Bakker
MA
,
Groen
HJ
,
Smit
EF
, et al
Chemotherapy for pulmonary large cell neuroendocrine carcinomas: does the regimen matter?
Eur Respir J
2017
;
49
:
160183
.
15.
Naidoo
J
,
Santos-Zabala
ML
,
Lyriboz
T
,
Woo
KM
,
Sima
CS
,
Fiore
JJ
, et al
Large cell neuroendocrine carcinoma of the lung: clinico-pathologic features, treatment, and outcomes
.
Clin Lung Cancer
2016
;
17
:
e121
9
.
16.
Christopoulos
P
,
Engel-Riedel
W
,
Grohe
C
,
Kropf-Sanchen
C
,
von Pawel
J
,
Gutz
S
, et al
Everolimus with paclitaxel and carboplatin as first-line treatment for metastatic large-cell neuroendocrine lung carcinoma: a multicenter phase II trial
.
Ann Oncol
2017
;
28
:
1898
902
.
17.
Rekhtman
N
,
Pietanza
MC
,
Hellmann
M
,
Naidoo
J
,
Arora
A
,
Won
H
, et al
Next-generation sequencing of pulmonary large cell neuroendocrine carcinoma reveals small cell carcinoma-like and non-small cell carcinoma-like subsets
.
Clin Cancer Res
2016
;
22
:
3618
29
.
18.
George
J
,
Fernandez-Cuesta
L
,
Vonn
W
,
Hayes
N
,
Thomas
RK
. 
Comparative analysis of small cell lung cancer and other pulmonary neuroendocrine tumors
.
Cancer Res
2016
;
76
(
14 suppl
):122.
19.
Casparie
M
,
Tiebosch
AT
,
Burger
G
,
Blauwgeers
H
,
van de Pol
A
,
van Krieken
JH
, et al
Pathology databanking and biobanking in The Netherlands, a central role for PALGA, the nationwide histopathology and cytopathology data network and archive
.
Cell Oncol
2007
;
29
:
19
24
.
20.
Socinski
MA
,
Smit
EF
,
Lorigan
P
,
Konduri
K
,
Reck
M
,
Szczesna
A
, et al
Phase III study of pemetrexed plus carboplatin compared with etoposide plus carboplatin in chemotherapy-naive patients with extensive-stage small-cell lung cancer
.
J Clin Oncol
2009
;
27
:
4787
92
.
21.
Hou
J
,
Lambers
M
,
den Hamer
B
,
den Bakker
MA
,
Hoogsteden
HC
,
Grosveld
F
, et al
Expression profiling-based subtyping identifies novel non-small cell lung cancer subgroups and implicates putative resistance to pemetrexed therapy
.
J Thorac Oncol
2012
;
7
:
105
14
.
22.
Monica
V
,
Scagliotti
GV
,
Ceppi
P
,
Righi
L
,
Cambieri
A
,
Lo Iacono
M
, et al
Differential thymidylate synthase expression in different variants of large-cell carcinoma of the lung
.
Clin Cancer Res
2009
;
15
:
7547
52
.
23.
Ceppi
P
,
Volante
M
,
Ferrero
A
,
Righi
L
,
Rapa
I
,
Rosas
R
, et al
Thymidylate synthase expression in gastroenteropancreatic and pulmonary neuroendocrine tumors
.
Clin Cancer Res
2008
;
14
:
1059
64
.
24.
Rindi
G
,
Klersy
C
,
Inzani
F
,
Fellegara
G
,
Ampollini
L
,
Ardizzoni
A
, et al
Grading the neuroendocrine tumors of the lung: an evidence-based proposal
.
Endocr Relat Cancer
2014
;
21
:
1
16
.
25.
Li
H
,
Handsaker
B
,
Wysoker
A
,
Fennell
T
,
Ruan
J
,
Homer
N
, et al
The Sequence Alignment/Map format and SAMtools
.
Bioinformatics
2009
;
25
:
2078
9
.
26.
Fernandez-Cuesta
L
,
Perdomo
S
,
Avogbe
PH
,
Leblay
N
,
Delhomme
TM
,
Gaborieau
V
, et al
Identification of circulating tumor DNA for the early detection of small-cell lung cancer
.
EBioMedicine
2016
;
10
:
117
23
.
27.
Wang
K
,
Li
M
,
Hakonarson
H
. 
ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data
.
Nucleic Acids Res
2010
;
38
:
e164
.
28.
Sim
NL
,
Kumar
P
,
Hu
J
,
Henikoff
S
,
Schneider
G
,
Ng
PC
. 
SIFT web server: predicting effects of amino acid substitutions on proteins
.
Nucleic Acids Res
2012
;
40
:
W452
7
.
29.
Adzhubei
IA
,
Schmidt
S
,
Peshkin
L
,
Ramensky
VE
,
Gerasimova
A
,
Bork
P
, et al
A method and server for predicting damaging missense mutations
.
Nat Methods
2010
;
7
:
248
9
.
30.
Lek
M
,
Karczewski
KJ
,
Minikel
EV
,
Samocha
KE
,
Banks
E
,
Fennell
T
, et al
Analysis of protein-coding genetic variation in 60,706 humans
.
Nature
2016
;
536
:
285
91
.
31.
Exome Variant Server
.
NHLBI GO Exome Sequencing Project (ESP)
; 
2017
.
Available from
: http://evs.gs.washington.edu/EVS/.
32.
Abecasis
GR
,
Altshuler
D
,
Auton
A
,
Brooks
LD
,
Durbin
RM
,
Gibbs
RA
, et al
A map of human genome variation from population-scale sequencing
.
Nature
2010
;
467
:
1061
73
.
33.
George
J
,
Lim
JS
,
Jang
SJ
,
Cun
Y
,
Ozretic
L
,
Kong
G
, et al
Comprehensive genomic profiles of small cell lung cancer
.
Nature
2015
;
524
:
47
53
.
34.
Shapiro
GI
,
Edwards
CD
,
Kobzik
L
,
Godleski
J
,
Richards
W
,
Sugarbaker
DJ
, et al
Reciprocal Rb inactivation and p16INK4 expression in primary lung cancers and cell lines
.
Cancer Res
1995
;
55
:
505
9
.
35.
Beasley
MB
,
Lantuejoul
S
,
Abbondanzo
S
,
Chu
WS
,
Hasleton
PS
,
Travis
WD
, et al
The P16/cyclin D1/Rb pathway in neuroendocrine tumors of the lung
.
Hum Pathol
2003
;
34
:
136
42
.
36.
Le Treut
J
,
Sault
MC
,
Lena
H
,
Souquet
PJ
,
Vergnenegre
A
,
Le Caer
H
, et al
Multicentre phase II study of cisplatin-etoposide chemotherapy for advanced large-cell neuroendocrine lung carcinoma: the GFPC 0302 study
.
Ann Oncol
2013
;
24
:
1548
52
.
37.
Niho
S
,
Kenmotsu
H
,
Sekine
I
,
Ishii
G
,
Ishikawa
Y
,
Noguchi
M
, et al
Combination chemotherapy with irinotecan and cisplatin for large-cell neuroendocrine carcinoma of the lung: a multicenter phase II study
.
J Thorac Oncol
2013
;
8
:
980
4
.
38.
Miyoshi
T
,
Umemura
S
,
Matsumura
Y
,
Mimaki
S
,
Tada
S
,
Ishii
G
, et al
Genomic profiling of large-cell neuroendocrine carcinoma of the lung
.
Clin Cancer Res
2016
;
23
:
757
65
.
39.
Karlsson
A
,
Brunnstrom
H
,
Lindquist
KE
,
Jirstrom
K
,
Jonsson
M
,
Rosengren
F
, et al
Mutational and gene fusion analyses of primary large cell and large cell neuroendocrine lung cancer
.
Oncotarget
2015
;
6
:
22028
37
.
40.
Simbolo
M
,
Mafficini
A
,
Sikora
KO
,
Fassan
M
,
Barbi
S
,
Corbo
V
, et al
Lung neuroendocrine tumours: deep sequencing of the four World Health Organization histotypes reveals chromatin-remodelling genes as major players and a prognostic role for TERT, RB1, MEN1 and KMT2D
.
J Pathol
2017
;
241
:
488
500
.
41.
Igarashi
T
,
Jiang
SX
,
Kameya
T
,
Asamura
H
,
Sato
Y
,
Nagai
K
, et al
Divergent cyclin B1 expression and Rb/p16/cyclin D1 pathway aberrations among pulmonary neuroendocrine tumors
.
Mod Pathol
2004
;
17
:
1259
67
.
42.
Dowlati
A
,
Lipka
MB
,
McColl
K
,
Dabir
S
,
Behtaj
M
,
Kresak
A
, et al
Clinical correlation of extensive-stage small-cell lung cancer genomics
.
Ann Oncol
2016
;
27
:
642
7
.
43.
Otterson
GA
,
Kratzke
RA
,
Coxon
A
,
Kim
YW
,
Kaye
FJ
. 
Absence of p16INK4 protein is restricted to the subset of lung cancer lines that retains wildtype RB
.
Oncogene
1994
;
9
:
3375
8
.
44.
Kwang Chae
Y
,
Tamragouri
K
,
Chung
J
,
Schrock
AB
,
Kolla
B
,
Ganesan
S
, et al
Genomic alterations (GA) and tumor mutational burden (TMB) in large cell neuroendocrine carcinoma of lung (L-LCNEC) as compared to small cell lung carcinoma (SCLC) as assessed via comprehensive genomic profiling (CGP)
.
J Clin Oncol
35
:15s, 
2017
(
suppl; abstr 8517
).