已入深夜,您辛苦了!由于当前在线用户较少,发布求助请尽量完整的填写文献信息,科研通机器人24小时在线,伴您度过漫漫科研夜!祝你早点完成任务,早点休息,好梦!

Single‐cell multi‐omics reveals tumor microenvironment factors underlying poor immunotherapy responses in ALK‐positive lung cancer

免疫疗法 肿瘤微环境 癌症研究 肺癌 生物 免疫系统 髓样 间变性淋巴瘤激酶 癌症 免疫检查点 癌症免疫疗法 医学 免疫学 肿瘤科 遗传学 恶性胸腔积液
作者
Seungbyn Baek,Euijeong Sung,Gamin Kim,Min Hee Hong,Chang Young Lee,Hyo Sup Shim,Sung Yong Park,Hye Ryun Kim,Insuk Lee
出处
期刊:Cancer communications [Wiley]
标识
DOI:10.1002/cac2.12658
摘要

Lung cancer remains the leading cause of cancer death in 2024, with ∼80% being non-small cell lung cancer (NSCLC). Anaplastic lymphoma kinase (ALK) rearrangements occur in ∼5% of NSCLC cases, typically treated with ALK inhibitors, though resistance often develops [1]. Immunotherapy has been explored for advanced or resistant ALK-positive NSCLC, but immune checkpoint blockade (ICB) treatments have shown limited clinical benefits [1]. A comprehensive study of ALK-positive NSCLC tumor microenvironment (TME) is needed to understand immunotherapy limitations and improve treatment strategies. We generated and collected single-cell RNA sequencing (scRNA-seq) and single-cell Assay for Transposase Accessible Chromatin with high-throughput sequencing (scATAC-seq) datasets from lung adenocarcinoma (LUAD) patients with ALK rearrangements and wild type without major oncogenic drivers (WT) (Supplementary Table S1). By comparing TME, we aimed to identify features explaining poor immunotherapy responses (Figure 1A). After batch corrections for each of the RNA and ATAC profiles, we identified epithelial, stromal, and immune cells (Supplementary Figure S1A, Supplementary Table S2). From the immune compartment, we identified major cell types such as myeloid cells, T cells, natural killer (NK) cells, and B cells (Figure 1B, Supplementary Figure S1B, Supplementary Table S3). Integration of the RNA and ATAC profiles confirmed consistency between these two omics profiles (Supplementary Figure S1C-D). Compositional analysis revealed that ALK-positive samples showed an enrichment of innate immune cells (myeloid and NK cells) and depletion of adaptive immune cells (T and B cells) (Figure 1C, Supplementary Figure S1E-F), suggesting weak adaptive anti-tumoral responses in ALK-positive TME. Oncogenic mutations primarily impact epithelial cells, so we classified epithelial cell malignancy with the cell-classifier and copy number variations (Supplementary Figure S1G-H) and measured ALK and PD-L1 expression. Malignant cells expressing ALK or PD-L1 were predominantly from ALK-positive malignant cells (Supplementary Figure S1I). We observed that ALK-positive tumors had a higher malignant-to-normal epithelial cell ratio than WT tumors (Figure 1D). Additionally, malignant cells in ALK-positive tumors exhibited increased stemness, indicating greater developmental potential (Figure 1E). Top 100 upregulated genes in ALK-positive tumors were linked to aggressive cancer pathways like epithelial-mesenchymal transition (EMT) and hypoxia [2] (Figure 1F). To identify malignant subsets with higher progression potential, we constructed developmental trajectories (Supplementary Figure S1J). State 4, enriched with ALK-positive malignant cells, showed higher stemness and elevated stress, hypoxia, and EMT gene signatures (Supplementary Figure S1J-L, Supplementary Table S4). Survival analysis with LUAD patients in The Cancer Genome Atlas (TCGA) [3] showed worse outcomes for patients with higher expression of State 4 signature genes, while differentially expressed genes (DEGs) between ALK-positive and WT malignant cells did not predict survival (Supplementary Figure S1M). This suggested that ALK rearrangement leads to more aggressive and metastatic tumors and emergence of clinically unfavorable malignant sub-states. Cell-cell interaction analysis showed ALK-positive malignant cells had increased interactions with myeloid cells (Figure 1G). Among these myeloid cell subtypes, ALK-positive tumors had significantly higher proportions of tumor-associated macrophages (TAMs) (Supplementary Figure S2A). ALK-positive TAMs were linked to M2-like pro-tumoral functions like adipogenesis and lipid metabolism, while WT TAMs were associated with M1-like immune-related functions, including TNF-alpha signaling and interferon gamma response (Figure 1H). Sub-clustering of TAMs (Supplementary Figure S2B-C) and RNA velocity analysis revealed WT TAMs transitioned into anti-tumoral, inflammation-related states, whereas ALK-positive TAMs transitioned into pro-tumoral, metabolic states (Figure 1I-J, Supplementary Figure S2D), maintaining immunosuppressive TME in ALK-positive tumors. Depleted adaptive immunity in ALK-positive tumors suggested reduced anti-tumoral immune functions [4]. B cell analysis (Supplementary Figure S2E-F) revealed that memory B cells and plasma cells were more abundant in WT tumors (Figure 1K). Top DEGs of WT memory B cells were enriched in oxidative phosphorylation pathways, indicating functional activation and effector capabilities [5] (Supplementary Figure S2G). Reconstructed B cell receptor sequences showed a lower ratio of expanded to non-expanded memory B cells in ALK-positive tumors (Figure 1L), suggesting reduced humoral anti-tumoral responses and activation. B cells are key components of tertiary lymphoid structures (TLSs), vital for immunotherapy responses [6]. We found positive correlations between B cells and CD4+ T cells, with WT tumors having more B cells (Figure 1M). ALK-positive tumors showed reduced interactions among major components of TLS, suggesting fewer TLS-like structures (Figure 1N), contributing to inadequate anti-tumoral responses and unfavorable TME for immunotherapy. Further experimental validations would be beneficial to confirm the existence of TLS-like structures. For CD8+ T cells, crucial in ICB treatment, we identified subtypes using marker genes (Supplementary Figure S2H-I). DEG analysis showed tumor-reactive and exhaustion genes (CXCL13, ENTPD1, ITGAE) were enriched in WT tumors, while bystander-indicating NK receptors (KLRK1, KLRC2) [7] were enriched in ALK-positive tumors (Figure 1O). T cell signature analysis confirmed these findings (Figure 1P, Supplementary Table S4). This suggested that ALK-positive "exhausted" CD8+ T cells became dysfunctional through non-canonical mechanisms, reducing their effectiveness during ICB treatment. Single-cell T cell receptor (TCR) sequencing showed fewer expanded ALK-positive CD8+ T cells with smaller TCR clonal sizes (Supplementary Figure S2J-K), indicating reduced T cell antigenicity. DEG analysis between expanded and non-expanded effector and exhausted CD8+ T cells revealed WT tumors had increased expression of exhaustion and tumor-reactivity genes, while the ALK-positive tumors showed decreased expression of these genes and increased NK receptor expression (Figure 1Q). This reaffirmed impaired tumor reactivity and more bystander-like CD8+ T cells in ALK-positive tumors. We analyzed gene regulatory changes in exhausted CD8+ T cell networks due to ALK rearrangement (Supplementary Figure S2L-M). In the WT network, T cell activation and exhaustion-related genes emerged as hubs with IFNG as a top hub gene, consistent with its role in effector T cell stimulation and reported deficiency in ALK-positive patients [1] (Supplementary Figure S2N). NK receptors like KLRK1 had high centrality in the ALK-positive network. We re-evaluated hub genes based on neighboring gene expression signatures. IFNG signature score was higher in exhausted T cells of WT tumors, while KLRK1 signature score was higher in exhausted T cells of ALK-positive tumors (Figure 1R). Since T cell exhaustion is also epigenetically regulated, we identified CD8+ T cell subtypes using scATAC-seq profiles and marker gene scores (Supplementary Figure S2O-P). Although transcription of exhaustion and tumor-reactivity genes was downregulated in ALK-positive CD8+ T cells, no clear depletion in chromatin accessibility was observed (Supplementary Figure S2Q), suggesting transcription factor (TF) binding caused these differences. Motif analysis showed BATF-related TFs, linked to CD8+ T cell effector functions [8, 9], were enriched in WT, while ETS1, important for NK cell differentiation [10], was enriched in ALK-positive cells (Figure 1S). We hypothesized that BATF is a key TF contributing to differences in T cell dysfunction. Using DEGs in CD8+ T cells between WT and ALK-positive tumors (see Figure 1O), we found WT DEGs had stronger BATF motif enrichment (Figure 1T) and all exhaustion/tumor-reactivity genes, except KRT86, had at least one peak with BATF motifs (Figure 1U). Interestingly, KRT86, the only exhaustion-related gene upregulated in expanded cells from both groups (see Figure 1Q), lacked BATF motifs. These findings suggested that differences in TF activities could lead to differences in gene expression. Additionally, WT exhausted T cells had more nearby peaks containing BATF motifs with higher co-accessibility, suggesting stronger co-regulation with nearby genomic regions in WT compared to the ALK-positive tumors (Figure 1V). In summary, this study provides key insights into the TME of ALK-positive lung cancer through single-cell multi-omics analysis, shedding light on poor immunotherapy responses. ALK-positive tumors displayed aggressive malignant phenotypes, enriched pro-tumoral myeloid cells, and depleted adaptive T and B cells. ALK-positive TAMs shifted away from inflammatory states, while CD8+ T cells showed reduced tumor reactivity and more bystander traits. We suggest that these CD8+ T cells may have distinct dysfunction mechanisms. Epigenetic analysis revealed depleted BATF motifs in ALK-positive CD8+ T cells, indicating altered TF activity. These findings highlight how ALK rearrangements drive an immunosuppressive TME, hindering effective immune responses, and suggest a need for strategies to reinvigorate adaptive immunity in ALK-positive lung cancer. Hye Ryun Kim, Seong Yong Park, and Insuk Lee conceived the study. Seungbyn Baek performed single-cell multi-omics data analysis under the supervision of Insuk Lee. Euijeong Sung assisted bioinformatic analysis. Gamin Kim contributed sample preparation. Hye Ryun Kim and Seong Yong Park organized clinical sample and data collections. Min Hee Hong and Chang Young Lee contributed to clinical sample collection. Hyo Sup Shim contributed to the pathological examination of tumor tissues. Insuk Lee and Hye Ryun Kim contributed to the financial and administrative support for this study. Seungbyn Baek, Hye Ryun Kim, and Insuk Lee wrote the manuscript. All authors read and approved the final manuscript. Not applicable. The authors declare that they have no conflicts of interest. This research was supported by the Bio & Medical Technology Development Program of the National Research Foundation funded by the Ministry of Science and ICT (2021R1A2C2094629 and 2017M3A9E9072669 to Hye Ryun Kim, and 2018R1A5A2025079, 2022M3A9F3016364, and 2022R1A2C1092062 to Insuk Lee). The work was supported in part by Brain Korea 21 (BK21) FOUR program. This work was supported by the Technology Innovation Program (20022947) funded by the Ministry of Trade Industry & Energy (MOTIE, Korea). This work was supported by the Yonsei Fellow Program, funded by Lee Youn Jae. The studies were approved by the Institutional Review Board of Yonsei University Severance Hospital with IRB No 4-2018-1161-1. Written informed consent was obtained prior to enrollment and sample collection at Yonsei University Severance Hospital. The research conformed to the principles of the Helsinki Declaration. The single-cell RNA, TCR, and ATAC sequencing data generated in this study are deposited in the Gene Expression Omnibus database with accession number GSE274934 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?&acc=GSE274934). The data will be available upon publication. The remaining data are available within the Article or Supplementary Information. 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.

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
一条鱼在北京完成签到,获得积分10
1秒前
流星完成签到,获得积分10
2秒前
2秒前
2秒前
5秒前
6秒前
Lucas应助aaa采纳,获得30
7秒前
缥缈映安完成签到 ,获得积分10
7秒前
pugongying完成签到,获得积分10
9秒前
9秒前
9秒前
天天快乐应助虚拟的凡波采纳,获得10
10秒前
sk夏冰完成签到 ,获得积分10
12秒前
科研的狗完成签到,获得积分10
12秒前
14秒前
14秒前
星际舟发布了新的文献求助10
15秒前
st完成签到,获得积分10
15秒前
16秒前
李D完成签到,获得积分10
16秒前
bkagyin应助科研通管家采纳,获得10
17秒前
Daphne发布了新的文献求助10
17秒前
17秒前
ding应助科研通管家采纳,获得10
18秒前
脑洞疼应助科研通管家采纳,获得10
18秒前
Akim应助科研通管家采纳,获得30
18秒前
Ava应助科研通管家采纳,获得10
18秒前
上官雨时发布了新的文献求助10
19秒前
aaa发布了新的文献求助30
21秒前
marshyyy发布了新的文献求助10
23秒前
李爱国应助suxin采纳,获得10
27秒前
27秒前
xzy998应助微笑飞鸟采纳,获得10
29秒前
无名老大应助aaa采纳,获得30
29秒前
31秒前
32秒前
等等发布了新的文献求助10
35秒前
无聊完成签到,获得积分10
37秒前
完美世界应助张大鹅采纳,获得10
37秒前
爆米花应助yao采纳,获得10
40秒前
高分求助中
Licensing Deals in Pharmaceuticals 2019-2024 3000
Cognitive Paradigms in Knowledge Organisation 2000
Mantiden: Faszinierende Lauerjäger Faszinierende Lauerjäger Heßler, Claudia, Rud 1000
PraxisRatgeber: Mantiden: Faszinierende Lauerjäger 1000
Natural History of Mantodea 螳螂的自然史 1000
A Photographic Guide to Mantis of China 常见螳螂野外识别手册 800
How Maoism Was Made: Reconstructing China, 1949-1965 800
热门求助领域 (近24小时)
化学 医学 材料科学 生物 工程类 有机化学 生物化学 物理 内科学 纳米技术 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 量子力学 冶金 电极
热门帖子
关注 科研通微信公众号,转发送积分 3319184
求助须知:如何正确求助?哪些是违规求助? 2950435
关于积分的说明 8551448
捐赠科研通 2627453
什么是DOI,文献DOI怎么找? 1437744
科研通“疑难数据库(出版商)”最低求助积分说明 666404
邀请新用户注册赠送积分活动 652388