单细胞多组学揭示了alk阳性肺癌免疫治疗反应差的肿瘤微环境因素。

IF 20.1 1区 医学 Q1 ONCOLOGY
Seungbyn Baek, Euijeong Sung, Gamin Kim, Min Hee Hong, Chang Young Lee, Hyo Sup Shim, Seong Yong Park, Hye Ryun Kim, Insuk Lee
{"title":"单细胞多组学揭示了alk阳性肺癌免疫治疗反应差的肿瘤微环境因素。","authors":"Seungbyn Baek,&nbsp;Euijeong Sung,&nbsp;Gamin Kim,&nbsp;Min Hee Hong,&nbsp;Chang Young Lee,&nbsp;Hyo Sup Shim,&nbsp;Seong Yong Park,&nbsp;Hye Ryun Kim,&nbsp;Insuk Lee","doi":"10.1002/cac2.12658","DOIUrl":null,"url":null,"abstract":"<p>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 [<span>1</span>]. Immunotherapy has been explored for advanced or resistant ALK-positive NSCLC, but immune checkpoint blockade (ICB) treatments have shown limited clinical benefits [<span>1</span>].</p><p>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).</p><p>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.</p><p>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 <i>ALK</i> and <i>PD-L1</i> expression. Malignant cells expressing <i>ALK</i> or <i>PD-L1</i> 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 [<span>2</span>] (Figure 1F).</p><p>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) [<span>3</span>] 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.</p><p>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.</p><p>Depleted adaptive immunity in ALK-positive tumors suggested reduced anti-tumoral immune functions [<span>4</span>]. 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 [<span>5</span>] (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 [<span>6</span>]. We found positive correlations between B cells and CD4<sup>+</sup> 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.</p><p>For CD8<sup>+</sup> T cells, crucial in ICB treatment, we identified subtypes using marker genes (Supplementary Figure S2H-I). DEG analysis showed tumor-reactive and exhaustion genes (<i>CXCL13, ENTPD1, ITGAE</i>) were enriched in WT tumors, while bystander-indicating NK receptors (<i>KLRK1, KLRC2</i>) [<span>7</span>] 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<sup>+</sup> T cells became dysfunctional through non-canonical mechanisms, reducing their effectiveness during ICB treatment.</p><p>Single-cell T cell receptor (TCR) sequencing showed fewer expanded ALK-positive CD8<sup>+</sup> 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<sup>+</sup> 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<sup>+</sup> T cells in ALK-positive tumors.</p><p>We analyzed gene regulatory changes in exhausted CD8<sup>+</sup> 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 <i>IFNG</i> as a top hub gene, consistent with its role in effector T cell stimulation and reported deficiency in ALK-positive patients [<span>1</span>] (Supplementary Figure S2N). NK receptors like <i>KLRK1</i> had high centrality in the ALK-positive network. We re-evaluated hub genes based on neighboring gene expression signatures. <i>IFNG</i> signature score was higher in exhausted T cells of WT tumors, while <i>KLRK1</i> signature score was higher in exhausted T cells of ALK-positive tumors (Figure 1R).</p><p>Since T cell exhaustion is also epigenetically regulated, we identified CD8<sup>+</sup> 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<sup>+</sup> 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<sup>+</sup> T cell effector functions [<span>8, 9</span>], were enriched in WT, while ETS1, important for NK cell differentiation [<span>10</span>], was enriched in ALK-positive cells (Figure 1S).</p><p>We hypothesized that BATF is a key TF contributing to differences in T cell dysfunction. Using DEGs in CD8<sup>+</sup> 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 <i>KRT86</i>, had at least one peak with BATF motifs (Figure 1U). Interestingly, <i>KRT86</i>, 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).</p><p>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<sup>+</sup> T cells showed reduced tumor reactivity and more bystander traits. We suggest that these CD8<sup>+</sup> T cells may have distinct dysfunction mechanisms. Epigenetic analysis revealed depleted BATF motifs in ALK-positive CD8<sup>+</sup> 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.</p><p>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.</p><p>The authors declare that they have no conflicts of interest.</p><p>This research was supported by the Bio &amp; 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 &amp; Energy (MOTIE, Korea). This work was supported by the Yonsei Fellow Program, funded by Lee Youn Jae.</p><p>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.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"45 4","pages":"422-427"},"PeriodicalIF":20.1000,"publicationDate":"2025-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12658","citationCount":"0","resultStr":"{\"title\":\"Single-cell multi-omics reveals tumor microenvironment factors underlying poor immunotherapy responses in ALK-positive lung cancer\",\"authors\":\"Seungbyn Baek,&nbsp;Euijeong Sung,&nbsp;Gamin Kim,&nbsp;Min Hee Hong,&nbsp;Chang Young Lee,&nbsp;Hyo Sup Shim,&nbsp;Seong Yong Park,&nbsp;Hye Ryun Kim,&nbsp;Insuk Lee\",\"doi\":\"10.1002/cac2.12658\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>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 [<span>1</span>]. Immunotherapy has been explored for advanced or resistant ALK-positive NSCLC, but immune checkpoint blockade (ICB) treatments have shown limited clinical benefits [<span>1</span>].</p><p>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).</p><p>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.</p><p>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 <i>ALK</i> and <i>PD-L1</i> expression. Malignant cells expressing <i>ALK</i> or <i>PD-L1</i> 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 [<span>2</span>] (Figure 1F).</p><p>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) [<span>3</span>] 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.</p><p>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.</p><p>Depleted adaptive immunity in ALK-positive tumors suggested reduced anti-tumoral immune functions [<span>4</span>]. 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 [<span>5</span>] (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 [<span>6</span>]. We found positive correlations between B cells and CD4<sup>+</sup> 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.</p><p>For CD8<sup>+</sup> T cells, crucial in ICB treatment, we identified subtypes using marker genes (Supplementary Figure S2H-I). DEG analysis showed tumor-reactive and exhaustion genes (<i>CXCL13, ENTPD1, ITGAE</i>) were enriched in WT tumors, while bystander-indicating NK receptors (<i>KLRK1, KLRC2</i>) [<span>7</span>] 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<sup>+</sup> T cells became dysfunctional through non-canonical mechanisms, reducing their effectiveness during ICB treatment.</p><p>Single-cell T cell receptor (TCR) sequencing showed fewer expanded ALK-positive CD8<sup>+</sup> 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<sup>+</sup> 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<sup>+</sup> T cells in ALK-positive tumors.</p><p>We analyzed gene regulatory changes in exhausted CD8<sup>+</sup> 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 <i>IFNG</i> as a top hub gene, consistent with its role in effector T cell stimulation and reported deficiency in ALK-positive patients [<span>1</span>] (Supplementary Figure S2N). NK receptors like <i>KLRK1</i> had high centrality in the ALK-positive network. We re-evaluated hub genes based on neighboring gene expression signatures. <i>IFNG</i> signature score was higher in exhausted T cells of WT tumors, while <i>KLRK1</i> signature score was higher in exhausted T cells of ALK-positive tumors (Figure 1R).</p><p>Since T cell exhaustion is also epigenetically regulated, we identified CD8<sup>+</sup> 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<sup>+</sup> 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<sup>+</sup> T cell effector functions [<span>8, 9</span>], were enriched in WT, while ETS1, important for NK cell differentiation [<span>10</span>], was enriched in ALK-positive cells (Figure 1S).</p><p>We hypothesized that BATF is a key TF contributing to differences in T cell dysfunction. Using DEGs in CD8<sup>+</sup> 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 <i>KRT86</i>, had at least one peak with BATF motifs (Figure 1U). Interestingly, <i>KRT86</i>, 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).</p><p>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<sup>+</sup> T cells showed reduced tumor reactivity and more bystander traits. We suggest that these CD8<sup>+</sup> T cells may have distinct dysfunction mechanisms. Epigenetic analysis revealed depleted BATF motifs in ALK-positive CD8<sup>+</sup> 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.</p><p>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.</p><p>The authors declare that they have no conflicts of interest.</p><p>This research was supported by the Bio &amp; 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 &amp; Energy (MOTIE, Korea). This work was supported by the Yonsei Fellow Program, funded by Lee Youn Jae.</p><p>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.</p>\",\"PeriodicalId\":9495,\"journal\":{\"name\":\"Cancer Communications\",\"volume\":\"45 4\",\"pages\":\"422-427\"},\"PeriodicalIF\":20.1000,\"publicationDate\":\"2025-01-04\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12658\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer Communications\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12658\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12658","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

肺癌仍然是2024年癌症死亡的主要原因,其中约80%为非小细胞肺癌(NSCLC)。间变性淋巴瘤激酶(ALK)重排发生在约5%的非小细胞肺癌病例中,通常用ALK抑制剂治疗,尽管耐药性经常发生。免疫疗法已被用于晚期或耐药alk阳性NSCLC,但免疫检查点阻断(ICB)治疗显示出有限的临床益处[10]。需要对alk阳性NSCLC肿瘤微环境(TME)进行全面的研究,以了解免疫治疗的局限性和改进治疗策略。我们生成并收集了来自ALK重排和无主要致癌驱动因素(WT)的野生型肺腺癌(LUAD)患者的单细胞RNA测序(scRNA-seq)和转座酶可及染色质高通量测序(scATAC-seq)单细胞检测数据集(补充表S1)。通过比较TME,我们旨在确定解释免疫治疗不良反应的特征(图1A)。在对每个RNA和ATAC谱进行批量校正后,我们鉴定了上皮细胞、基质细胞和免疫细胞(补充图S1A,补充表S2)。从免疫区室,我们确定了主要的细胞类型,如骨髓细胞、T细胞、自然杀伤(NK)细胞和B细胞(图1B,补充图S1B,补充表S3)。RNA和ATAC图谱的整合证实了这两个组学图谱之间的一致性(补充图S1C-D)。成分分析显示,alk阳性样本显示先天免疫细胞(髓细胞和NK细胞)富集,适应性免疫细胞(T细胞和B细胞)耗竭(图1C,补充图S1E-F),表明alk阳性TME的适应性抗肿瘤反应较弱。致癌突变主要影响上皮细胞,因此我们通过细胞分类器和拷贝数变化对上皮细胞恶性进行分类(Supplementary Figure S1G-H),并测量ALK和PD-L1的表达。表达ALK或PD-L1的恶性细胞主要来自ALK阳性的恶性细胞(补充图S1I)。我们观察到alk阳性肿瘤的恶性上皮细胞与正常上皮细胞的比例高于WT肿瘤(图1D)。此外,alk阳性肿瘤中的恶性细胞表现出更高的干性,表明更大的发育潜力(图1E)。alk阳性肿瘤中的前100个上调基因与侵袭性癌症途径相关,如上皮-间质转化(EMT)和缺氧[2](图1F)。为了确定具有更高发展潜力的恶性亚群,我们构建了发展轨迹(补充图S1J)。状态4富含alk阳性恶性细胞,表现出更高的干性,应激、缺氧和EMT基因特征升高(补充图S1J-L,补充表S4)。在癌症基因组图谱(TCGA)[3]中对LUAD患者的生存分析显示,State 4特征基因高表达的患者预后较差,而alk阳性和WT恶性细胞之间的差异表达基因(DEGs)不能预测生存(补充图S1M)。这表明ALK重排导致肿瘤更具侵袭性和转移性,并出现临床不利的恶性亚状态。细胞间相互作用分析显示,alk阳性恶性细胞与髓系细胞的相互作用增加(图1G)。在这些髓系细胞亚型中,alk阳性肿瘤中肿瘤相关巨噬细胞(tam)的比例明显更高(补充图S2A)。alk阳性的tam与m2样的促肿瘤功能相关,如脂肪生成和脂质代谢,而WT tam与m1样的免疫相关功能相关,包括tnf - α信号传导和干扰素γ反应(图1H)。tam的亚聚类(补充图S2B-C)和RNA速度分析显示,WT tam转变为抗肿瘤、炎症相关状态,而alk阳性tam转变为促肿瘤、代谢状态(图1 - j,补充图S2D),在alk阳性肿瘤中维持免疫抑制的TME。alk阳性肿瘤的适应性免疫缺失提示抗肿瘤免疫功能降低。B细胞分析(Supplementary Figure S2E-F)显示,记忆性B细胞和浆细胞在WT肿瘤中更为丰富(Figure 1K)。WT记忆B细胞的顶部DEGs在氧化磷酸化途径中富集,表明功能激活和效应能力[5](补充图S2G)。重建的B细胞受体序列显示,在alk阳性肿瘤中,扩展记忆B细胞与非扩展记忆B细胞的比例较低(图1L),表明体液抗肿瘤反应和激活降低。B细胞是三级淋巴样结构(TLSs)的关键组成部分,对免疫治疗反应至关重要。 我们发现B细胞与CD4+ T细胞呈正相关,WT肿瘤中B细胞较多(图1M)。alk阳性肿瘤显示TLS主要组分之间的相互作用减少,表明TLS样结构较少(图1N),导致抗肿瘤反应不足,不利于免疫治疗的TME。进一步的实验验证将有助于证实类tls结构的存在。对于在ICB治疗中至关重要的CD8+ T细胞,我们使用标记基因确定了亚型(补充图S2H-I)。DEG分析显示,肿瘤反应性和衰竭基因(CXCL13, ENTPD1, ITGAE)在WT肿瘤中富集,而旁观者指示NK受体(KLRK1, KLRC2)[7]在alk阳性肿瘤中富集(图10)。T细胞特征分析证实了这些发现(图1P,补充表S4)。这表明alk阳性的“耗尽”CD8+ T细胞通过非规范机制变得功能失调,降低了它们在ICB治疗期间的有效性。单细胞T细胞受体(TCR)测序显示,扩增的alk阳性CD8+ T细胞较少,TCR克隆大小较小(Supplementary Figure S2J-K),表明T细胞抗原性降低。扩增和非扩增效应和耗尽的CD8+ T细胞之间的DEG分析显示,WT肿瘤的耗尽和肿瘤反应性基因表达增加,而alk阳性肿瘤的这些基因表达减少,NK受体表达增加(图1Q)。这再次确认了alk阳性肿瘤中受损的肿瘤反应性和更多的旁观者样CD8+ T细胞。我们分析了ALK重排导致的耗尽CD8+ T细胞网络的基因调控变化(补充图S2L-M)。在WT网络中,T细胞激活和耗竭相关基因作为枢纽出现,IFNG作为顶部枢纽基因,这与IFNG在效应T细胞刺激中的作用和alk阳性患者[1]中报道的缺陷一致(补充图S2N)。NK受体如KLRK1在alk阳性网络中具有较高的中心性。我们根据邻近基因表达特征重新评估枢纽基因。WT肿瘤的衰竭T细胞中IFNG特征评分较高,而alk阳性肿瘤的衰竭T细胞中KLRK1特征评分较高(图1R)。由于T细胞耗竭也受表观遗传调控,我们使用scATAC-seq谱和标记基因评分鉴定了CD8+ T细胞亚型(补充图s20 - p)。尽管在alk阳性CD8+ T细胞中,耗竭基因和肿瘤反应性基因的转录下调,但没有观察到染色质可及性的明显减少(补充图S2Q),这表明转录因子(TF)的结合导致了这些差异。基序分析显示,与CD8+ T细胞效应功能相关的batf相关tf[8,9]在WT中富集,而对NK细胞分化[10]重要的ETS1在alk阳性细胞中富集(图1S)。我们假设BATF是导致T细胞功能障碍差异的关键TF。在WT和alk阳性肿瘤之间的CD8+ T细胞中使用DEGs(见图10),我们发现WT DEGs具有更强的BATF基元富集(图1T),并且除KRT86外,所有衰竭/肿瘤反应性基因至少具有一个BATF基元峰(图1U)。有趣的是,KRT86是两组扩增细胞中唯一上调的衰竭相关基因(见图1Q),缺乏BATF基元。这些结果表明,TF活性的差异可能导致基因表达的差异。此外,WT耗尽的T细胞有更多含有BATF基序的附近峰,具有更高的共可及性,这表明与alk阳性肿瘤相比,WT与附近基因组区域的共调节更强(图1V)。总之,本研究通过单细胞多组学分析为alk阳性肺癌的TME提供了关键见解,揭示了免疫治疗反应不良的原因。alk阳性肿瘤表现为侵袭性恶性表型,肿瘤前骨髓细胞丰富,适应性T细胞和B细胞减少。alk阳性的tam从炎症状态转移,而CD8+ T细胞表现出肿瘤反应性降低和更多的旁观者特征。我们认为这些CD8+ T细胞可能有不同的功能障碍机制。表观遗传学分析显示,alk阳性CD8+ T细胞中BATF基序缺失,表明TF活性改变。这些发现强调了ALK重排如何驱动免疫抑制性TME,阻碍有效的免疫应答,并提示需要重新激活ALK阳性肺癌的适应性免疫策略。Hye Ryun Kim、Seong Yong Park和Insuk Lee构思了这项研究。Seungbyn Baek在Insuk Lee的指导下进行了单细胞多组学数据分析。Euijeong Sung协助进行生物信息学分析。Gamin Kim贡献了样品制备。Hye Ryun Kim和Seong Yong Park组织了临床样本和数据收集。 Min Hee Hong和Chang Young Lee为临床样本收集做出了贡献。Hyo Sup Shim对肿瘤组织的病理检查有贡献。Insuk Lee和Hye Ryun Kim为这项研究提供了财政和行政支持。白承彬、金惠润、李仁淑等人撰写了手稿。所有作者都阅读并批准了最终的手稿。作者声明他们没有利益冲突。这项研究得到了Bio &amp;科学和信息通信技术部国家研究基金医疗技术发展计划(20121r1a2c2094629和2017M3A9E9072669资助给Hye Ryun Kim, 2018R1A5A2025079, 2022M3A9F3016364和2022R1A2C1092062资助给Insuk Lee)。这项工作得到了Brain Korea 21 (BK21) FOUR项目的部分支持。本研究受国家工贸部科技创新计划(20022947)资助;能源(MOTIE,韩国)。这项工作得到了延世研究员计划的支持,由Lee Youn Jae资助。这些研究得到了延世大学Severance医院机构审查委员会(IRB No . 4-2018-1161-1)的批准。在延世大学Severance医院登记和采集样本前获得书面知情同意。这项研究符合《赫尔辛基宣言》的原则。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

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

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

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.

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.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信