Molecular subtypes of SCLC and their clinical relevance
In the respective analysis of the two patient cohorts and cell lines, SCLC was classified into four subtypes: SCLC-A of ASCL1+, SCLC-I of CD274+ (PD-L1), SCLC-N of NEUROD1+, and SCLC-P of POU2F3+ (Fig. 1A and B). We verified the targeted panel expression profile of the NCC cohort and compared it with that of the whole transcriptome in the George et al. dataset 13. The two datasets exhibited high correlation coefficients with the four subtypes (R > 0.73; Fig. 1A). Three master regulators, ASCL1, NEUROD1, and POU2F3, were upregulated in the three subtypes SCLC-A/N/P (Fig. 1B and Supplementary Fig. S1). SCLC-I was observed more frequently in the NCC cohort (29.9%) than in the George et al. cohort (17.3%), whereas SCLC-N showed the opposite trend (NCC, 14.9%; George et al. 33.3%; Supplementary Table S1). In terms of clinical profiles, the NCC cohort comprised 80.5% of ED (stage IV) cases, and the George et al. cohort comprised 55.6% of ED cases. In terms of the clinical profile of NCC, the SCLC-A group was the youngest, and the SCLC-P group was the oldest (P = 0.01; Fig. 1C and Supplementary Table S1). Brain metastasis (NCC, 14.9%) was more frequently observed in the SCLC-I (OR = 1.57) and SCLC-P (OR = 1.71; Fig. 1C) groups.
Pathological investigation of the master regulators further validated the performance of our molecular subtype classification (Fig. 1C). We performed immunohistochemical (IHC) analysis for various markers and assessed regulation of the signature target genes bound by the three master regulators. The IHC showed that ASCL1+ was enriched in SCLC-A and SCLC-I/N. POU2F3 clearly defined only the SCLC-P subtype (SCLC-P, P = 3.89e-04; Fig. 1C). Moreover, NEUROD1 IHC staining was non-specific and failed to classify SCLC-N. Additionally, TTF1+ was relatively abundant, except in SCLC-P. MYC+ was abundant in SCLC-I and SCLC-P. CD56 was a positive marker for all SCLC samples.
Differentially expressed genes (DEGs) were used to define the distinct regulatory programs for each subtype. The expression of known marker genes was consistent with the expected results (P < 6.0e-05; Supplementary Fig. S1). ASCL1, DLL3, FOXA1, and SOX2 were overexpressed in SCLC-A cells. Wnt signaling and FOXA1/3 transcription factor network pathways were enriched in SCLC-A (Fig. 1D, Supplementary Table S2). CD274 (PD-L1) and T-cell receptor (TCR) signaling were activated in SCLC-I. High NE scores were observed for both SCLC-A and SCLC-N. The mitotic cell cycle was dysregulated in both SCLC-A/N cell lines. PI3K-Akt signaling was activated in SCLC-N. POU2F3 and MYC were dramatically upregulated in SCLC-P cells. Notch signaling and extracellular matrix (ECM) organization contributed to the SCLC-P regulatory program.
Early developmental status of SCLC identified from TP53/RB1-mutated NSCLC resembles that of SCLC-A
Both TP53 and RB1 variants are key drivers of SCLC development5,13. The histological transformation from NSCLC to SCLC confers therapeutic challenges in sharing TP53/RB1 variants5. To define the early developmental status of SCLC in NSCLC, we explored whether the transcriptome profile of NSCLC accompanies the subtype features of SCLC. Accordingly, we investigated the gene expression profiles of samples harboring TP53/RB1 variants from The Cancer Genome Atlas (TCGA) LUAD (n = 510) and squamous cell carcinoma (lung squamous cell carcinoma (LUSC); n = 484) datasets. We extracted data for the LUADTP53/RB1 (n = 4) and LUSCTP53/RB1 (n = 7) samples.
ASCL1 was significantly upregulated in LUADTP53/RB1 samples compared with that in wild-type LUADs (P = 0.02); however, no difference was observed in NEUROD1 (P = 0.5) and POU2F3 (P = 0.83; Fig. 2A). In LUSC, NEUROD1 was upregulated in LUSC TP53/RB1 (P = 0.05, Fig. 2A). NE scores were significantly higher in LUADTP53/RB (P = 0.0004) than wild-type LUAD, but not in LUSCTP53/RB (P = 0.59; Fig. 2B). We also analyzed the expression of SCLC DEG regulators in LUAD. The SCLC-A regulators FOXA1 and SOX2 were upregulated in LUADTP53/RB (P < 0.003). In contrast, the genes of the remaining three subtypes SCLC-I/N/P (CD274, MYC, NOTCH2, REST, and SOX9) showed no difference in expression (Fig. 2C). Upon comparing the global gene expression and NE scores of LUAD TP53/RB with those of SCLC subtypes, LUAD TP53/RB expression was found to be close to that for SCLC-A and SCLC-N (Fig. 2D and E). Additionally, LUADTP53/RB showed a trend toward worse progression-free survival (PFS) (hazard ratio [HR] = 2.65; P = 0.08) and overall survival (OS) (P = 0.058). Based on these findings, we concluded that LUADTP53/RB exhibits SCLC NE gene expression, and its molecular features are similar to those of the SCLC-A subtype.
Single-cell clusters to dissect endothelial-to-mesenchymal transition (EndMT) phenotype in SCLC-I type
Although NE-type cell development is well characterized, the SCLC TME is not well defined. A previous study involving immune cell analysis revealed cytotoxic T-cell infiltration in SCLC-I and a dysregulated immune response by exhausted T cells in SCLC-N 7. However, the TME cell types and their evolution are not fully understood. Therefore, we next investigated the single-cell transcriptome to select non-NE cells to identify TME cells. We also examined TME cell subclusters and clonal evolution. To characterize TME cells, we acquired a single-cell transcriptome profile of patient malignant cells (treatment-naïve: 7 patients, 6,585 cells; previously treated: 10 patients, 5,927 cells; as described in Materials and methods) 7. Previously treated patients were administered platinum-based cytotoxic agents (etoposide, cisplatin, or carboplatin) and immune checkpoint inhibitors (ipilimumab or atezolizumab).
We classified NE and non-NE cells and clustered the SCLC TME cells with non-NE cells. POU2F3+ staining was observed among the subclusters of non-NE cells. Interestingly, post-treatment SCLC cells comprised 14.2% non-NE cells, whereas treatment-naïve cells comprised 1.1% non-NE cells (Fig. 3A). Therefore, we selected non-NE cells from the post-treatment samples. Compared with NE cells, which exhibit ASCL1 and TFF3 upregulation (P < 1.42e-111), heterogenous non-NE cells presented IFITM3, B2M, ANXA4, VIM, CD74, S100A11, and YAP1 overexpression (P < 5.77e-57, fold change (FC) > 1; Supplementary Fig. S2A). Interferon signaling, cell cycle, and antigen processing were simultaneously activated in non-NE cells (Supplementary Fig. S2B). NE cells exhibited ASCL1, TFF3, and DLL3 upregulation (Supplementary Fig. S2A and B).
Non-NE cells, mostly TME cells, were extracted from all previously treated patients without any sample bias (Supplementary Fig. S2C). Subsequently, non-NE cells were classified into five clusters (Fig. 3B; Supplementary Fig. S2C). Five clusters were annotated using overexpression markers: low-NE, myeloid, epithelial, T-cell, and endothelial (Fig. 3B; P < 1.27e-05; Supplementary Fig. S2D; Supplementary Tables S3 and S4). ASCL1 was relatively active in low-NE cells to maintain cell cycle activation and P53 signal dysregulation (P < 6.18e-04; Supplementary Fig. S2D; Supplementary Table S5). POU2F3 upregulation was observed in the epithelial cluster (P = 4.62e-15; Supplementary Fig. S2E). Myeloid cells also exhibited MKI67, CDK1, and TOP2A overexpression, contributing to an aberrant cell cycle (Supplementary Table S5). The T-cell cluster was identified using PTPRC (CD45) staining. Additional T-cell subtypes were evaluated using IHC on the NCC cohort samples. IFN-γ and TCR staining were detected for T-cell subtype genes. Furthermore, the endothelial clusters were found to modulate the ECM, angiogenesis, and vascular endothelial growth factor (VEGF) signaling. The epithelial cluster showed enrichment in the biological processes of cell adhesion, interferon-alpha/beta, and TGF-beta signaling. These results suggest that VIM+ cancer-associated fibroblasts (CAF) features govern endothelial and epithelial clusters. These two clusters were considered the previously defined EndMT and POU2F3+ epithelial-to-mesenchymal transition (EMT) clusters, respectively.
The non-NE SCLC population evolved into distinct TME cell types. The five TME cell clusters expanded from low-NE cells to three distinct branches: myeloid, T, and endo/epithelial cells (Fig. 3C). We further investigated the abundance of the five TME cell types among the the SCLC subtypes. Low-NE cells were enriched in SCLC-A (P = 2.28e-07; Fig. 3D). Endothelial and T-cell signatures were abundant in SCLC-I (P < 7.0e-04). The remaining cell types were not significantly different among the four SCLC subtypes. Moreover, we verified T-cell infiltration using an mIHC/IF-based assay in the NCC cohort (Fig. 3E, Supplementary Fig. S3). CD4+ and CD8+ T cells were abundant in SCLC-I, as were CD4+/FOXP3, CD8+/PD-L1, and CD20 (P < 0.06; S18-26664; Kruskal–Wallis H test, P < 0.004; Fig. 3F, Supplementary Fig. S3). Non-NE cells showed an apparent increase in number after treatment and evolved into three branches: myeloid, T, and endo/epithelial cells. Specifically, SCLC-I comprised a heterogeneous TME to facilitate EndMT and CD8+/PD-L1 T-cell infiltration.
SCLC TME cell cluster signatures to predict the worst outcome
Prognostic features of SCLC have been demonstrated in both restricted cohorts and mouse models. Here, we referred to two cohorts to explore the genetic regulations promoting poor outcomes. First, ED patients were found to have poor survival compared with limited disease (LD) patients in terms of both 2-year PFS (P = 0.07, HR = 1.85) and OS (P = 0.01, HR = 2.65) in NCC (Fig. 4A). The PFS and OS according to LD/ED status were not significant in the George et al. cohort. However, the non-NE-type clearly distinguished poor outcomes between the two cohorts (PFS George et al., P = 0.09; NCC, P = 0.1; Fig. 4B).
Additionally, we performed survival tests for TME molecular features (Supplementary Table S3). Survival, according to the four SCLC subtypes, failed to reach statistical significance. However, the regulatory programs reflected by the five TME subclusters were prognostic factors. In the George et al. cohort, T-cell infiltration was associated with a better prognosis (Fig. 4C). Compared with patients with T-cell activation, endothelial signal-activated patients exhibited worse outcomes (P = 0.1, HR = 1.84). CD4, CD8, and Treg IHC staining for the NCC cohort was similar to that for the George et al. cohort (Supplementary Fig. S4). Because only genes of three signatures were available in the NCC-targeted panel, we referred to myeloid signaling for comparison. Endothelial signature indicated the worst outcome (P = 0.14, HR = 1.73; Fig. 4C, bottom panel). We presumed that the TME heterogeneity of SCLC-I encompasses contradictory prognostic factors. Even with T-cell infiltration, the endothelial signature indicated the worst outcome in the SCLC-I subtype.
SCLC molecular features and drug candidates for resistance to platinum-based chemotherapy
To characterize the initial sensitivity and rapid progression of SCLC to chemotherapy, we referred to the cisplatin response scores analyzed from the SCLC transcriptome profile. SCLC-A and SCLC-N were sensitive, whereas SCLC-I was the most resistant (P = 6.84e-06; Fig. 5A)14. Among the TME cell types, the endothelial and T-cell-activated groups also participated in cisplatin resistance (P = 1.79e-07; Fig. 5A). This implied that TME development in the SCLC-I subtype was associated with cisplatin resistance, whereas the two NE subtypes remained sensitive to cisplatin.
We evaluated cisplatin resistance scores from high-throughput drug screening data for cell lines and PDCs. The drug response values of the SCLC cell lines also resembled cisplatin response scores (Fig. 5B). Unfortunately, the IC50 values of cisplatin and etoposide are unavailable for Cancer Cell Line Encyclopedia (CCLE)15. However, the NE-type cells were consistently sensitive to platinum-based drugs (oxaliplatin; FC = 0.34, P = 0.04) and histone deacetylase (HDAC) inhibitors (MS-275 and SB939; FC > 0.28, P < 0.09). Moreover, the endothelial-activated cell types were sensitive to bromodomain and extra-terminal motif (BET) inhibitors (JQ1 and GSK-1210151A; FC > 0.55, P < 0.05) and a graft inhibitor (pelitrexol; FC = 0.43, P < 0.08). To cross-evaluate the CCLE results, we investigated additional drug response screening profiles acquired for 366 PDCs and 64 drugs (Supplementary Table S6). Upon comparing SCLC (n = 21) with NSCLC (n = 341), drugs targeting Chk1, Aurora, Wee1, and PI3K-Akt were identified as candidates for SCLC sensitivity. However, the comparison between cisplatin-sensitive and -resistant groups within SCLC indicated different candidates compared with NSCLC. Cell cycle target drugs were no longer predicted to be effective against cisplatin-resistant SCLC PDCs. Cisplatin-resistant PDCs exhibited concurrent resistance to barasertib, alisertib (Aurora), camptothecin (CPT; topoisomerase), and everolimus (mTOR; FC<-1.26, P < 0.04). Effective drugs for cisplatin-resistant cells were barely identified with a strict P value cutoff. Brigatinib (ALK/EGFR inhibitor), dasatinib (multiple TKI), and JQ1 (BET inhibitor) were considered potential candidates based on FC (FC > 0.61), even though the P value cutoff was not reached. Our results imply that the response to chemotherapy resembled that to cell cycle-targeting drugs or other cytotoxic drugs.
Finally, we performed an integrative analysis of the co-expressed regulatory target genes based on the candidate JQ1 response. Upon assessing highly correlated genes affected by JQ1 treatment (negative IC50), we observed enrichment of the extracellular matrix organization, Hippo, WNT, and angiogenesis pathways (P < 3.85e-03, Supplementary Table S6). CCND1 was the top-ranked factor involved in the Hippo and WNT signaling pathways (R = 0.7, P = 9.94e-08; Fig. 5D). TGIF2 and SMAD3 in the TGF-β signaling pathway were also highly correlated (R > 0.59, P < 0.002). Other correlated pathways were also associated with EndMT. JQ1, a BET inhibitor targeting BRD4, suppresses angiogenesis in various cancer types16. As reported previously, EndMT in non-NE cell types increases platinum resistance. To overcome platinum resistance, our data propose BET inhibitors as novel therapeutic candidates to suppress angiogenesis activation in SCLC-I.