Abstract

Small cell lung cancer (SCLC) features with high heterogeneity and poor prognosis. The major treatment strategy for SCLC remains combination chemotherapy over past decades, with only minor improvements.1 And the application of immunotherapy has no impressive benefit for SCLC patients.2 In this study, we identify a novel subtype of SCLC (SCLC-I) with immunosuppressive feature and high genomic instability. Importantly, we find that POU2F3 is effective in predicting SCLC-I, and the POU2F3-high patients exhibit better responses to immunotherapy. We performed weighted correlation network analysis (WGCNA)3 to explore heterogeneity of SCLC through integrating transcriptomic data detailed in the Supplementary Material (SM), including 19 Chinese surgical samples and 112 samples from two public data.4, 5 We firstly transformed gene expression data into co-expression module to capture genes with similar expression pattern and extracted 17 coherent modules. All SCLC samples were clustered into four subtypes (Figure 1A), which showed no obvious difference in survival (Figure 1B). Each subtype showed comparable percentages in these three datasets (Figure 1A, Figure S5A), indicative of equal contribution of these datasets in the integrative analysis. To compare our findings with the previous study, in which SCLC was classified into SCLC-A, SCLC-N, SCLC-P and SCLC-Y,6 we evaluated the relative expression of these markers in our clusters. We observed that cluster 2 and cluster 3 corresponded to the SCLC-A and SCLC-N, respectively (Figure 1C). Consistent with the observation from Gay et al.,7 we found that YAP1 had low expression and did not exclusively define a subtype of SCLC (Figure 1C). We found that CCSP (SCGB1A1), a secreted protein mainly produced by club cells to maintain airway integrity,8 was specifically highly expressed in cluster 4 (Figure 1E, Figure S3E), indicating that certain SCLC might originate from club cells. Hereafter, we named cluster 4 as SCLC-C. The cluster 1 showed low ASCL1 and NEUROD1 expression and high POU2F3 and NOTCH2 expression (Figure 1C,D). Gene set enrichment analysis (GSEA) demonstrated that the interferon-gamma response, interferon-alpha response, inflammatory response and IL-6/JAK/STAT3 signaling were enriched in SCLC-I subtype (Figure 1F), which clearly confirmed its immune-related characteristic. We therefore named cluster 1 as 'immune subtype (SCLC-I)'. Further analysis showed that the enrichment of immune-related pathways, such as IL-10 signaling and other interleukin-related pathways, in SCLC-I versus other subtypes (Figure 2A, Figure S4B). Multiple immuno-inhibitory factors such as PD1, IL-10, IDO1, CD96 and BTLA, were highly expressed in SCLC-I (Figure 2B,E, Figure S4). Immune cell infiltration is considered to be primary immune signature and strongly associated with the clinical outcome of cancer immunotherapies.9 Using ImmuCellAI,10 we found that the abundance of dendritic cells, macrophage, induced regulatory T cells (iTreg) and CD8+ T cells were significantly increased in SCLC-I (Figure 2C). Moreover, we observed that most of the chemokines, such as CXCL10, CCL17, CCL18, were up-regulated in SCLC-I subtype (Figure 2D). Taken together, SCLC-I subtype is featured with the activation of immune checkpoint molecules and infiltration of immune suppressive cells such as iTreg, which might help the immune escape. To explore the unique genetic alterations of each subtype, we performed analyses of 115 samples with available genomic sequencing data and RNA-sequencing data. We found that each subtype harbored specific gene mutations, for example, LRP1, CD163, MME, ABCB1 mutations were frequently observed in SCLC-I (Figure 3A). In comparison with other samples, SCLC-I group showed significantly higher genetic alterations (Figure 3B,C), which indicated the increased genomic instability. SCLC-I had more gene amplifications on Chr2, Chr6, Chr11, Chr12 and Chr19, whereas SCLC-A had more gene amplifications on Chr17, SCLC-N had more amplified genes on Chr9 and Chr21, and SCLC-C had more amplified genes on Chr14 (Figure 3E, Table S2). Over 3000 genes were observed significantly amplified with high alteration frequency (>50%) in SCLC-I (Figure 3D, Table S2). Further analysis showed that the cholesterol biosynthesis I pathway was significantly enriched in SCLC-I subtype (Figure 3F). Consistent with the GSEA result using gene expression profile (Figure 1F), the WNT/beta-catenin signaling was enriched in both SCLC-I and SCLC-N subtypes. Collectively, these data show that SCLC-I subtype has higher level of genomic instability and each subtype harbors unique gene mutations and copy number variations. To determine whether our findings have clinical relevance, we then identified the biomarker for SCLC-I. According to feature selection from random forest model, we found that 10 genes (POU2F3, ANXA1, LRMP, GFI1B, SLC7A14, PHYHIPL, MAP2, SYP, KCNK3 and CPE) were most important in distinguishing SCLC-I from other subtypes (Figure 4A, SM 6 and Figure S6A). Using these genes to build random forest model on 100 sets of different testing data, we got the average prediction accuracy of 92.74% and average score of area under curve (AUC) of 93.32% (Figure 4B, Table S3). Among 10 genes used for SCLC-I prediction, POU2F3 stood out as the most significantly up-regulated gene with high expression (Figures 4C and 1C and Table S5). Moreover, the SCLC-P samples identified in the previous study6 were all included in SCLC-I with up-regulated immune-related pathways (Figure S6C). We further collected a cohort containing 28 relapsed SCLC samples from patients receiving immunotherapy or chemo-immunotherapy (Table S4, SM 7) and performed immunohistochemical staining of POU2F3 in these specimens (Figure 4D, Table S4). Our data showed that the patients with high POU2F3 expression exhibited a significantly improved objective response rate (ORR) to immunotherapy, with a high AUC of 0.813 (Figure 4E). Moreover, the POU2F3 protein level was positively correlated with patient prognosis (Figure 4F). Importantly, two patients with high POU2F3 level showed dramatic regression of lung tumours (Figure 4G,H). The positive response to immunotherapy indicated the potentially strong immune cell infiltration in POU2F3-high SCLC. These results together supported that the SCLC-I patients are more sensitive to immunotherapy, and POU2F3 might serve as a biomarker for SCLC immunotherapy. In conclusion, our work systematically uncovers the transcriptomic and genomic heterogeneity in SCLC and characterizes a novel immune subtype with high sensitivity to immunotherapy. We identified POU2F3 as a potential biomarker with a good prediction power to assess SCLC immunotherapy response. Gay et al. identifies an inflamed subtype of SCLC which shows a significant overall survival (OS) benefit relative to all other subtypes with the combined chemotherapy and immunotherapy.7 In our study, the immune subtype seems correspond to a combination of SCLC-P and SCLC-I from Gay et al. cohorts. Although we used a different method and biomarkers to identify this special subtype of SCLC, both our study and Gay et al. study have proven the potential of SCLC re-clustering in current clinical immunotherapy. Of course, the small size of the validation cohort is a limiting factor. Future clinical efforts and larger cohorts are required to validate the effectiveness of immunotherapy in this subtype. This work was supported by the National Basic Research Program of China (grant numbers: 2017YFA0505500 to Luonan Chen and Hongbin Ji and 2020YFA0803300 to Hongbin Ji), the Strategic Priority Research Program of the Chinese Academy of Sciences (grant numbers: XDB19020201 to Hongbin Ji and XDB38040400 to Luonan Chen), the National Natural Science Foundation of China (grant numbers: 31930022 and 31771476 to Luonan Chen; 82030083, 31621003, 81872312, and 82011540007 to Hongbin Ji; 81871875 to Liang Hu), Basic Frontier Scientific Research Program of Chinese Academy of Science (grant number: ZDBS-LY-SM006 to Hongbin Ji), International Cooperation Project of Chinese Academy of Sciences (grant number: 153D31KYSB20190035 to Hongbin Ji), Innovative research team of high-level local universities in Shanghai (grant number: SSMU-ZLCX20180500 to Hongbin Ji) and Science and Technology Commission of Shanghai Municipality (grant numbers: 15XD1504000 Hongbin Ji and 2017SHZDZX01 to Luonan Chen). The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.

Highlights

  • Small cell lung cancer (SCLC) is one of the deadliest cancers marked with fast proliferation and early metastasis

  • We identified four dominant subtypes with a robust clustering approach based on gene modules, two of which correspond well with the classic (ASCL1-high, NEUROD1-low) and variant (ASCL1-low, NEUROD1-high) subtypes

  • The remaining subtype represents a novel variant subtype characterized with lower expression of both ASCL1 and NEUROD1, higher expression of NOTCH1/2, showing increased immunogenic and consequent immunosuppressive features, which is defined as an immune subtype

Read more

Summary

Introduction

Small cell lung cancer (SCLC) is one of the deadliest cancers marked with fast proliferation and early metastasis. Two subtypes of SCLC have been previously recognized for decades: the classic subtype and the variant subtype[1, 7, 8, 9]. The lineage-specific transcription factor, ASCL1, is associated with the NE property and overexpressed in the classical subtype[10]. Down-regulation of ASCL1 expression in the variant subtype may contribute to its loss of NE property. Another transcription factor, NEUROD1, is reported to be important in maintenance of the variant subtype[10, 11]. Distinct four SCLC molecular subtypes have been proposed based on the expression of ASCL1, NEUROD1, POU2F3 and YAP112. The network-based subtyping with a larger dataset is expected to provide reliable subtypes of SCLC patients, in contrast to the four subtypes obtained by a traditional expression-based subtyping with a relatively small dataset[12]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call