Background: The tumor immune microenvironment of anaplastic lymphoma kinase (ALK)-rearranged lung adenocarcinoma (LUAD) stratified by ALK fusion variants is poorly pictured. Hence, in this study, we aim to explore the immune heterogeneity of ALK+ LUAD across different ALK fusion variants and further investigate their significance on clinical prognosis.
Methods: A retrospective analysis was conducted on ALK+ LUAD patients (N=68). DNA and RNA-based next-generation sequencing (NGS) was performed to clarify the specific ALK fusion variants. Clinical and pathological characteristics were compared between long and short ALK variants. To research the immune heterogeneity, multi-fluorescence was carried out to explore the differences in immune properties, such as tumor-infiltrating lymphocyte (TIL) number, TIL subset, and tertiary lymphoid structures (TLS) development, between long and short ALK variants. Furthermore, the prognostic value of these characteristics was analyzed. Finally, the expression of lymphocyte-activation gene-3 (LAG3), one novel immune therapy target, was assessed across ALK+ LUAD.
Results: LUAD patients with short ALK fusion variant-driven tumors exhibited higher American Joint Committee on Cancer (AJCC) stage as well as larger tumor size than those with long ALK fusion variant-driven tumors. Compared to long ALK fusion variants, there were more TILs, especially natural killer (NK) cells, within short ALK variants. However, fewer TLS were established in cancers harboring short ALK variants than those with long ALK variants. In advanced-stage LUAD patients with ALK fusion, short ALK variants, hot immune status, and high-level NK cells were identified to be adverse prognostic factors, while high-level B cells, as well as the development of TLS, served as positive prognostic factors. As for LAG3 expression, LAG3+ immune cells were more enriched in short ALK variants than in long ALK variants.
Conclusions: LUAD patients with short ALK fusion variant-driven tumors exhibited worse prognosis than those with long ALK fusion variant-driven tumors. The tumor immune microenvironments are heterogeneous across different ALK fusion variants with short variants characterized by higher levels of TIL, especially NK cells, but by less TLS development than long variants ALK+ LUAD, which disfavor disease outcomes.
{"title":"Characteristics of the immune microenvironment and their clinical significance in lung adenocarcinoma patients with different ALK fusion variants.","authors":"Yinbo Xiao, Hao Wang, Junliang Lu, Junyi Pang, Shiyi Liu, Yang Zhou, Xiaohua Shi, Zhiyong Liang","doi":"10.21037/tlcr-24-682","DOIUrl":"10.21037/tlcr-24-682","url":null,"abstract":"<p><strong>Background: </strong>The tumor immune microenvironment of anaplastic lymphoma kinase (ALK)-rearranged lung adenocarcinoma (LUAD) stratified by ALK fusion variants is poorly pictured. Hence, in this study, we aim to explore the immune heterogeneity of ALK<sup>+</sup> LUAD across different ALK fusion variants and further investigate their significance on clinical prognosis.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on ALK<sup>+</sup> LUAD patients (N=68). DNA and RNA-based next-generation sequencing (NGS) was performed to clarify the specific ALK fusion variants. Clinical and pathological characteristics were compared between long and short ALK variants. To research the immune heterogeneity, multi-fluorescence was carried out to explore the differences in immune properties, such as tumor-infiltrating lymphocyte (TIL) number, TIL subset, and tertiary lymphoid structures (TLS) development, between long and short ALK variants. Furthermore, the prognostic value of these characteristics was analyzed. Finally, the expression of lymphocyte-activation gene-3 (LAG3), one novel immune therapy target, was assessed across ALK<sup>+</sup> LUAD.</p><p><strong>Results: </strong>LUAD patients with short ALK fusion variant-driven tumors exhibited higher American Joint Committee on Cancer (AJCC) stage as well as larger tumor size than those with long ALK fusion variant-driven tumors. Compared to long ALK fusion variants, there were more TILs, especially natural killer (NK) cells, within short ALK variants. However, fewer TLS were established in cancers harboring short ALK variants than those with long ALK variants. In advanced-stage LUAD patients with ALK fusion, short ALK variants, hot immune status, and high-level NK cells were identified to be adverse prognostic factors, while high-level B cells, as well as the development of TLS, served as positive prognostic factors. As for LAG3 expression, LAG3<sup>+</sup> immune cells were more enriched in short ALK variants than in long ALK variants.</p><p><strong>Conclusions: </strong>LUAD patients with short ALK fusion variant-driven tumors exhibited worse prognosis than those with long ALK fusion variant-driven tumors. The tumor immune microenvironments are heterogeneous across different ALK fusion variants with short variants characterized by higher levels of TIL, especially NK cells, but by less TLS development than long variants ALK<sup>+</sup> LUAD, which disfavor disease outcomes.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3538-3554"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012002","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tlcr-24-326
Martin Reck, Konstantinos Syrigos, Skaidrius Miliauskas, Susan C Van't Westeinde, Bartomeu Massuti, Hannes Buchner, Alexey V Salnikov, Robert M Lorence, Anne-Marit Ellingboe, Thomas Kitzing, Keith Kerr
Background: Anti-angiogenic agents, such as nintedanib and ramucirumab, when combined with docetaxel, are subsequent treatment options in patients with non-small cell lung cancer (NSCLC) who have failed on first-line chemotherapy or immunochemotherapy. However, to date, there are no validated predictive biomarkers for efficacy of anti-angiogenic therapies in this setting. The aim of this study was to explore whether genetic or genomic markers, alone or combined with clinical covariates, could be used to predict overall survival (OS) in patients with NSCLC who are eligible for treatment with nintedanib plus docetaxel.
Methods: LUME-BioNIS (NCT02671422) was a prospective, non-interventional study that assessed the efficacy and safety of nintedanib plus docetaxel in patients with relapsed/refractory NSCLC. The primary outcome was OS in relation to exploratory molecular biomarkers, alone or in combination with clinical covariates. Exploratory multivariate and univariate analyses were undertaken on putative biomarkers including clinical variables, somatic mutations, gene expression, immunological, and proliferation markers. Sub-analyses in patients with prior immunotherapy were performed.
Results: Of 260 enrolled patients, most patients received nintedanib plus docetaxel in the second-line (68.8%) or third-line (25.8%). After a median follow-up of 19.7 months, median OS was 8.1 months (95% confidence interval: 7.1-9.5). Univariate subgroup analysis indicated that the presence of liver/adrenal metastases, Eastern Cooperative Oncology Group performance status (ECOG PS) ≥1, time since start of first-line therapy (<9 months), and response to first-line therapy had potential prognostic significance for OS. In multivariate analysis, the presence of brain/liver metastases and the presence of >2 metastatic sites at baseline were associated with OS. In univariate analyses in patients with prior immunotherapy, RNA expression levels of genes involved in cell proliferation, DNA damage repair, interferon signaling, and abundance of neutrophils had potential prognostic significance for OS.
Conclusions: Nintedanib plus docetaxel had promising activity and manageable safety in a real-world clinical setting. No new predictive biomarkers were identified to help select patients who may particularly benefit from anti-angiogenic therapy.
{"title":"A non-interventional biomarker study in patients with adenocarcinoma of the lung treated with nintedanib plus docetaxel following progression on chemotherapy and/or immunotherapy: LUME-BioNIS.","authors":"Martin Reck, Konstantinos Syrigos, Skaidrius Miliauskas, Susan C Van't Westeinde, Bartomeu Massuti, Hannes Buchner, Alexey V Salnikov, Robert M Lorence, Anne-Marit Ellingboe, Thomas Kitzing, Keith Kerr","doi":"10.21037/tlcr-24-326","DOIUrl":"https://doi.org/10.21037/tlcr-24-326","url":null,"abstract":"<p><strong>Background: </strong>Anti-angiogenic agents, such as nintedanib and ramucirumab, when combined with docetaxel, are subsequent treatment options in patients with non-small cell lung cancer (NSCLC) who have failed on first-line chemotherapy or immunochemotherapy. However, to date, there are no validated predictive biomarkers for efficacy of anti-angiogenic therapies in this setting. The aim of this study was to explore whether genetic or genomic markers, alone or combined with clinical covariates, could be used to predict overall survival (OS) in patients with NSCLC who are eligible for treatment with nintedanib plus docetaxel.</p><p><strong>Methods: </strong>LUME-BioNIS (NCT02671422) was a prospective, non-interventional study that assessed the efficacy and safety of nintedanib plus docetaxel in patients with relapsed/refractory NSCLC. The primary outcome was OS in relation to exploratory molecular biomarkers, alone or in combination with clinical covariates. Exploratory multivariate and univariate analyses were undertaken on putative biomarkers including clinical variables, somatic mutations, gene expression, immunological, and proliferation markers. Sub-analyses in patients with prior immunotherapy were performed.</p><p><strong>Results: </strong>Of 260 enrolled patients, most patients received nintedanib plus docetaxel in the second-line (68.8%) or third-line (25.8%). After a median follow-up of 19.7 months, median OS was 8.1 months (95% confidence interval: 7.1-9.5). Univariate subgroup analysis indicated that the presence of liver/adrenal metastases, Eastern Cooperative Oncology Group performance status (ECOG PS) ≥1, time since start of first-line therapy (<9 months), and response to first-line therapy had potential prognostic significance for OS. In multivariate analysis, the presence of brain/liver metastases and the presence of >2 metastatic sites at baseline were associated with OS. In univariate analyses in patients with prior immunotherapy, RNA expression levels of genes involved in cell proliferation, DNA damage repair, interferon signaling, and abundance of neutrophils had potential prognostic significance for OS.</p><p><strong>Conclusions: </strong>Nintedanib plus docetaxel had promising activity and manageable safety in a real-world clinical setting. No new predictive biomarkers were identified to help select patients who may particularly benefit from anti-angiogenic therapy.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3364-3381"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736584/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011972","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tlcr-24-659
Jihyun Park, Danbi Jo, Seo Yoon Choi, Sumin Oh, Yoon Seok Jung, Oh Yoen Kim, Juhyun Song
Background: Previous studies reported significant relationships between obesity and pulmonary dysfunction. Here, we investigated genetic alterations in the lung tissues of high fat diet (HFD) induced obese mouse through transcriptomic and molecular analyses.
Methods: Eight-week-old male C57BL/6J mice were fed either a normal chow diet (NCD) or HFD for 12 weeks. We performed RNA sequencing, functional analysis of altered genes using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway data, Database for Annotation, Visualization and Integrated Discovery (DAVID) analysis, protein network analysis, quantitative real-time polymerase chain reaction, and Western blotting.
Results: We performed RNA sequencing analysis in the lung tissue of HFD mice. GO and KEGG pathway data presented higher expressions of genes related to lung fibrosis, and the changes of several pathways including regulation of nitrogen compound metabolic process, G protein-coupled receptor signaling, cancer pathway, and small cell lung cancer pathway. DAVID analysis and protein network analysis showed the changes of vascular endothelial growth factor, hypoxia-inducible factor-1 and rat sarcoma virus signaling related to vascular permeability, and protein network of MYC proto-oncogene gene related to cancer. In addition, we found increased protein and mRNA levels of the growth/differentiation factor 15 and alpha smooth muscle actin genes related to lung fibrosis in lung tissue of HFD mice.
Conclusions: HFD contributes to an increased risk of lung fibrosis and lung cancer. Thus, we propose that the genetic modulation and the molecular regulation of target pathways are essential to suppress pulmonary fibrosis in obese patients.
{"title":"Long-term high fat diet aggravates the risk of lung fibrosis and lung cancer: transcriptomic analysis in the lung tissues of obese mice.","authors":"Jihyun Park, Danbi Jo, Seo Yoon Choi, Sumin Oh, Yoon Seok Jung, Oh Yoen Kim, Juhyun Song","doi":"10.21037/tlcr-24-659","DOIUrl":"https://doi.org/10.21037/tlcr-24-659","url":null,"abstract":"<p><strong>Background: </strong>Previous studies reported significant relationships between obesity and pulmonary dysfunction. Here, we investigated genetic alterations in the lung tissues of high fat diet (HFD) induced obese mouse through transcriptomic and molecular analyses.</p><p><strong>Methods: </strong>Eight-week-old male C57BL/6J mice were fed either a normal chow diet (NCD) or HFD for 12 weeks. We performed RNA sequencing, functional analysis of altered genes using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway data, Database for Annotation, Visualization and Integrated Discovery (DAVID) analysis, protein network analysis, quantitative real-time polymerase chain reaction, and Western blotting.</p><p><strong>Results: </strong>We performed RNA sequencing analysis in the lung tissue of HFD mice. GO and KEGG pathway data presented higher expressions of genes related to lung fibrosis, and the changes of several pathways including regulation of nitrogen compound metabolic process, G protein-coupled receptor signaling, cancer pathway, and small cell lung cancer pathway. DAVID analysis and protein network analysis showed the changes of vascular endothelial growth factor, hypoxia-inducible factor-1 and rat sarcoma virus signaling related to vascular permeability, and protein network of MYC proto-oncogene gene related to cancer. In addition, we found increased protein and mRNA levels of the growth/differentiation factor 15 and alpha smooth muscle actin genes related to lung fibrosis in lung tissue of HFD mice.</p><p><strong>Conclusions: </strong>HFD contributes to an increased risk of lung fibrosis and lung cancer. Thus, we propose that the genetic modulation and the molecular regulation of target pathways are essential to suppress pulmonary fibrosis in obese patients.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3513-3525"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736590/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Immune checkpoint inhibitor (ICI) therapy has prolonged the survival of a proportion of patients with advanced non-small cell lung cancer (NSCLC). Histological quantification of programmed cell death-ligand 1 (PD-L1) in tumors is a widely adopted marker for predicting the efficacy of ICI treatment. However, its use in patients with malignant pleural effusion (MPE) is occasionally challenging because of the difficulty of tissue sampling. The aim of this study was to identify new predictive factors for ICI treatment in patients with MPE.
Methods: A total of 22 patients with MPE were included. Initially, we surveyed several parameters of pleural effusion in relation to overall survival (OS). Next, we attempted to reflect the response to ICI treatment in vitro, a simple co-culture bioassay was designed, in which tumor cells and immune cells were co-cultured with nivolumab. Binding of nivolumab to T cells was confirmed by flow cytometry, and the released interferon-gamma (IFN-γ) was evaluated by enzyme-linked immunosorbent assay.
Results: The parameter analysis demonstrated that the levels of albumin and the percentage of lymphocyte were significantly correlated with OS in all patients. Vascular endothelial growth factor (VEGF) and high mobility group box 1 (HMGB1) were negatively correlated with OS in only driver gene mutation-positive patients. In vitro bioassay showed that nivolumab-binding T cells predominantly produced IFN-γ compared with control antibody. Of the 22 patients, 12 showed an increase in IFN-γ release after treatment with nivolumab. Despite the lack of significant correlations between IFN-γ levels and OS, the duration of ICI treatment tended to be longer in patients with IFN-γ release versus those without IFN-γ release.
Conclusions: This immune assay of IFN-γ release after treatment with nivolumab in vitro may identify responders prior to ICI treatment.
{"title":"A novel bioassay reflecting response to immune checkpoint inhibitor therapy in non-small cell lung cancer with malignant pleural effusion.","authors":"Ayako Takigami, Naoko Mato, Koichi Hagiwara, Makoto Maemondo","doi":"10.21037/tlcr-24-559","DOIUrl":"https://doi.org/10.21037/tlcr-24-559","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitor (ICI) therapy has prolonged the survival of a proportion of patients with advanced non-small cell lung cancer (NSCLC). Histological quantification of programmed cell death-ligand 1 (PD-L1) in tumors is a widely adopted marker for predicting the efficacy of ICI treatment. However, its use in patients with malignant pleural effusion (MPE) is occasionally challenging because of the difficulty of tissue sampling. The aim of this study was to identify new predictive factors for ICI treatment in patients with MPE.</p><p><strong>Methods: </strong>A total of 22 patients with MPE were included. Initially, we surveyed several parameters of pleural effusion in relation to overall survival (OS). Next, we attempted to reflect the response to ICI treatment <i>in vitro</i>, a simple co-culture bioassay was designed, in which tumor cells and immune cells were co-cultured with nivolumab. Binding of nivolumab to T cells was confirmed by flow cytometry, and the released interferon-gamma (IFN-γ) was evaluated by enzyme-linked immunosorbent assay.</p><p><strong>Results: </strong>The parameter analysis demonstrated that the levels of albumin and the percentage of lymphocyte were significantly correlated with OS in all patients. Vascular endothelial growth factor (VEGF) and high mobility group box 1 (HMGB1) were negatively correlated with OS in only driver gene mutation-positive patients. <i>In vitro</i> bioassay showed that nivolumab-binding T cells predominantly produced IFN-γ compared with control antibody. Of the 22 patients, 12 showed an increase in IFN-γ release after treatment with nivolumab. Despite the lack of significant correlations between IFN-γ levels and OS, the duration of ICI treatment tended to be longer in patients with IFN-γ release versus those without IFN-γ release.</p><p><strong>Conclusions: </strong>This immune assay of IFN-γ release after treatment with nivolumab <i>in vitro</i> may identify responders prior to ICI treatment.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3267-3277"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736611/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Differences in the immune microenvironment and responses to immunotherapy may exist between primary non-small cell lung cancer (NSCLC) and brain metastases (BMs). This study aimed to investigate discrepancies in programmed death-ligand 1 (PD-L1) expression, tumor-infiltrating lymphocytes (TILs), tertiary lymphoid structures (TLS), and tumor mutational burden (TMB) between matched BMs and primary tumors (PTs) in NSCLC.
Methods: Twenty-six pairs of surgically resected BMs and corresponding PTs from NSCLC patients were collected. PD-L1 expression and TILs, including CD8, CD3, CD4, CD20, CD68, and CD21, were analyzed using immunohistochemistry (IHC) and quantitatively assessed through digital image analysis. Whole-exome sequencing (WES) was performed to investigate genomic discrepancies and variations in TMB.
Results: The density of PD-L1+ cells did not differ significantly between matched PTs and BMs (P>0.99). However, BMs exhibited a higher tumor proportion score (TPS) compared to PTs (mean TPS: 31.92% vs. 25.96%, P=0.049), with moderate agreement in categorized TPS (κ=0.653). Analysis of TILs revealed a significant reduction in CD3+ T cells (P<0.001), CD8+ cytotoxic T cells (P<0.001), CD20+ B cells (P<0.001), and CD68+ macrophages (P=0.02) in BMs compared to PTs. BMs also exhibited a loss of TLS, with no presence of mature TLS marked by CD21 expression. The number of non-synonymous mutations was generally higher in BMs than in PTs, with only 34.69% of mutations shared between paired PTs and BMs. TMB was slightly increased in BMs (mean TMB: 34.2 mutations/Mb in BMs vs. 26.8 mutations/Mb in PTs; P=0.30). Additionally, the log-rank test indicated that a higher density of CD20+ B cells in BMs was significantly associated with better overall survival (P=0.007).
Conclusions: Compared to primary NSCLC tumors, matched BMs show an increase in TPS of PD-L1 expression and TMB, but a significant reduction in TILs and loss of mature TLS, suggesting an immune-suppressive microenvironment in BMs. The infiltration of CD20+ B cells may serve as a potential prognostic biomarker in NSCLC with BMs.
背景:原发性非小细胞肺癌(NSCLC)和脑转移瘤(BMs)在免疫微环境和免疫治疗反应方面可能存在差异。本研究旨在探讨非小细胞肺癌匹配脑转移瘤和原发肿瘤(PTs)在程序性死亡配体1 (PD-L1)表达、肿瘤浸润淋巴细胞(TILs)、三级淋巴结构(TLS)和肿瘤突变负荷(TMB)方面的差异。方法:收集非小细胞肺癌患者手术切除的脑转移灶及相应的PTs 26对。采用免疫组化(IHC)分析PD-L1表达和TILs,包括CD8、CD3、CD4、CD20、CD68和CD21,并通过数字图像分析定量评估。采用全外显子组测序(WES)研究TMB的基因组差异和变异。结果:PD-L1+细胞密度在匹配的PTs和脑转移瘤之间无显著差异(P < 0.99)。然而,脑转移患者的肿瘤比例评分(TPS)高于PTs(平均TPS: 31.92% vs. 25.96%, P=0.049),分类TPS的一致性中等(κ=0.653)。TILs分析显示,与PTs相比,脑转移灶中CD3+ T细胞(P+细胞毒性T细胞)(P+ B细胞)(P+巨噬细胞)显著减少(P=0.02)。脑转移瘤也表现出TLS缺失,不存在以CD21表达为标志的成熟TLS。脑转移患者的非同义突变数量普遍高于脑转移患者,只有34.69%的突变在配对的PTs和脑转移患者之间共享。脑转移患者的TMB略有增加(脑转移患者的平均TMB: 34.2个突变/Mb vs. PTs的26.8个突变/Mb;P = 0.30)。此外,log-rank检验表明,脑转移瘤中CD20+ B细胞密度越高,总生存率越高(P=0.007)。结论:与原发性NSCLC肿瘤相比,匹配的脑转移灶PD-L1表达的TPS和TMB增加,但TILs显著降低,成熟TLS缺失,提示脑转移灶存在免疫抑制微环境。CD20+ B细胞的浸润可能是伴有脑转移的非小细胞肺癌的潜在预后生物标志物。
{"title":"Discrepancies in PD-L1 expression, lymphocyte infiltration, and tumor mutational burden in non-small cell lung cancer and matched brain metastases.","authors":"Yanhui Zhang, Runfen Cheng, Tingting Ding, Jianghua Wu","doi":"10.21037/tlcr-24-735","DOIUrl":"https://doi.org/10.21037/tlcr-24-735","url":null,"abstract":"<p><strong>Background: </strong>Differences in the immune microenvironment and responses to immunotherapy may exist between primary non-small cell lung cancer (NSCLC) and brain metastases (BMs). This study aimed to investigate discrepancies in programmed death-ligand 1 (PD-L1) expression, tumor-infiltrating lymphocytes (TILs), tertiary lymphoid structures (TLS), and tumor mutational burden (TMB) between matched BMs and primary tumors (PTs) in NSCLC.</p><p><strong>Methods: </strong>Twenty-six pairs of surgically resected BMs and corresponding PTs from NSCLC patients were collected. PD-L1 expression and TILs, including CD8, CD3, CD4, CD20, CD68, and CD21, were analyzed using immunohistochemistry (IHC) and quantitatively assessed through digital image analysis. Whole-exome sequencing (WES) was performed to investigate genomic discrepancies and variations in TMB.</p><p><strong>Results: </strong>The density of PD-L1<sup>+</sup> cells did not differ significantly between matched PTs and BMs (P>0.99). However, BMs exhibited a higher tumor proportion score (TPS) compared to PTs (mean TPS: 31.92% <i>vs.</i> 25.96%, P=0.049), with moderate agreement in categorized TPS (κ=0.653). Analysis of TILs revealed a significant reduction in CD3<sup>+</sup> T cells (P<0.001), CD8<sup>+</sup> cytotoxic T cells (P<0.001), CD20<sup>+</sup> B cells (P<0.001), and CD68<sup>+</sup> macrophages (P=0.02) in BMs compared to PTs. BMs also exhibited a loss of TLS, with no presence of mature TLS marked by CD21 expression. The number of non-synonymous mutations was generally higher in BMs than in PTs, with only 34.69% of mutations shared between paired PTs and BMs. TMB was slightly increased in BMs (mean TMB: 34.2 mutations/Mb in BMs <i>vs.</i> 26.8 mutations/Mb in PTs; P=0.30). Additionally, the log-rank test indicated that a higher density of CD20<sup>+</sup> B cells in BMs was significantly associated with better overall survival (P=0.007).</p><p><strong>Conclusions: </strong>Compared to primary NSCLC tumors, matched BMs show an increase in TPS of PD-L1 expression and TMB, but a significant reduction in TILs and loss of mature TLS, suggesting an immune-suppressive microenvironment in BMs. The infiltration of CD20<sup>+</sup> B cells may serve as a potential prognostic biomarker in NSCLC with BMs.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3590-3602"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736585/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012136","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The role of pyroptosis in lung squamous cell carcinoma (LUSC) remains unclear. This study aimed to screen pyroptosis-related genes (PRGs) and construct a model to investigate the immune infiltration, gene mutations, and immune response of patients of LUSC.
Methods: We conducted a comprehensive evaluation of pyroptosis patterns in patients with LUSC with 51 PRGs. Pyroptosis-related clusters were identified using consistency clustering algorithm. Differences in the biologic and clinical characteristics between the clusters were analyzed. Cox regression analysis was performed to screen for differentially expressed genes (DEGs) related to prognosis, and a principal component analysis (PCA) algorithm was used to construct a model based on these genes. The pyroptosis score was calculated for each tumor sample, and the samples were classified into high- and low-score groups based on the score. The disparities in survival, single-nucleotide variation (SNV), copy number variation (CNV), and immunotherapy response between high-score and low-score groups were analyzed.
Results: A total of 51 PRGs were used to classify LUSC samples into three pyroptosis clusters with significant differences in survival (P=0.005). Based on the 390 DEGs between the three clusters, two distinct pyroptosis gene clusters were identified by secondary clustering, with significant differences in prognosis (P=0.005). A pyroptosis scoring model was established to evaluate the regulatory patterns of PRGs, and patients were stratified into two groups with high and low scores, using the median pyroptosis score as the cutoff. The survival analyses indicated that patients with high scores had worse prognoses in The Cancer Genome Atlas (TCGA)-LUSC cohort (P=0.002), which was further supported by the analysis of the GSE37745 (P=0.006) and GSE135222 datasets (P=0.02).
Conclusions: The quantification of pyroptosis patterns was found to be important in predicting prognosis and devising personalized treatment strategies in patients with LUSC.
{"title":"Identification and validation of pyroptosis patterns with a novel quantification system for the prediction of prognosis in lung squamous cell carcinoma.","authors":"Xianyu Qin, Jiayan Wu, Fei Qin, Yuzhen Zheng, Junguo Chen, Zui Liu, Jian Tan, Weijie Cai, Shiyun He, Bozhu Jian, Haosheng Zheng, Hongying Liao","doi":"10.21037/tlcr-24-1003","DOIUrl":"https://doi.org/10.21037/tlcr-24-1003","url":null,"abstract":"<p><strong>Background: </strong>The role of pyroptosis in lung squamous cell carcinoma (LUSC) remains unclear. This study aimed to screen pyroptosis-related genes (PRGs) and construct a model to investigate the immune infiltration, gene mutations, and immune response of patients of LUSC.</p><p><strong>Methods: </strong>We conducted a comprehensive evaluation of pyroptosis patterns in patients with LUSC with 51 PRGs. Pyroptosis-related clusters were identified using consistency clustering algorithm. Differences in the biologic and clinical characteristics between the clusters were analyzed. Cox regression analysis was performed to screen for differentially expressed genes (DEGs) related to prognosis, and a principal component analysis (PCA) algorithm was used to construct a model based on these genes. The pyroptosis score was calculated for each tumor sample, and the samples were classified into high- and low-score groups based on the score. The disparities in survival, single-nucleotide variation (SNV), copy number variation (CNV), and immunotherapy response between high-score and low-score groups were analyzed.</p><p><strong>Results: </strong>A total of 51 PRGs were used to classify LUSC samples into three pyroptosis clusters with significant differences in survival (P=0.005). Based on the 390 DEGs between the three clusters, two distinct pyroptosis gene clusters were identified by secondary clustering, with significant differences in prognosis (P=0.005). A pyroptosis scoring model was established to evaluate the regulatory patterns of PRGs, and patients were stratified into two groups with high and low scores, using the median pyroptosis score as the cutoff. The survival analyses indicated that patients with high scores had worse prognoses in The Cancer Genome Atlas (TCGA)-LUSC cohort (P=0.002), which was further supported by the analysis of the GSE37745 (P=0.006) and GSE135222 datasets (P=0.02).</p><p><strong>Conclusions: </strong>The quantification of pyroptosis patterns was found to be important in predicting prognosis and devising personalized treatment strategies in patients with LUSC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3657-3674"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736615/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-23DOI: 10.21037/tlcr-24-552
Magdalena Knetki-Wróblewska, Kamila Wojas-Krawczyk, Paweł Krawczyk, Maciej Krzakowski
Immune checkpoint inhibitors (ICIs) have become an established treatment option for patients with advanced non-small cell lung cancer (NSCLC). However, the efficacy of single-agent immunotherapy as well as in combination with chemotherapy seems to be dependent on the presence of molecular abnormalities in some genes-serine/threonine kinase 11 (STK11), Kelch-like ECH-associated protein 1 (KEAP1) and Kirsten rat sarcoma viral oncogene homolog (KRAS) among them. The KEAP1 gene is a critical regulator of the cellular response to oxidative stress and electrophilic stress, thus playing a pivotal role in maintaining cellular homeostasis. The STK11 gene encodes a serine/threonine kinase (STK11) involved the regulation of cell growth, polarity, motility, differentiation and cell metabolism. The STK11 gene mutations are often associated with an immunologically "cold" tumour microenvironment. The co-occurrence of STK11 or KEAP1 abnormalities with the KRAS mutation changes the composition of the tumour microenvironment as compared when presented alone. The current data, based on retrospective analyses of clinical trials, indicate that the co-existence of STK11 and KEAP1 genes mutations with the KRAS gene mutations have negative impact on the prognosis, regardless of treatment methods, in patients with advanced NSCLC. However, this group of patients should not be omitted because they constitute a significant percentage of advanced NSCLC patients. Immunotherapy focused on two ICIs [anti-programmed death 1 (PD-1)/anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4)] combined with chemotherapy, may be more effective than immunotherapy or chemotherapy alone in this group of patients. Confirmation of this thesis can be found in the results of available clinical studies. Here, we summarize the theoretical justification as well as the results of clinical trials for combining immunotherapy in patients with STK11-, KEAP1- and KRAS-mutated genes. There is certainly a need to create a prospective clinical trial to assess the effectiveness of combined immunotherapy in the discussed group of patients.
{"title":"Emerging insights into <i>STK11</i>, <i>KEAP1</i> and <i>KRAS</i> mutations: implications for immunotherapy in patients with advanced non-small cell lung cancer.","authors":"Magdalena Knetki-Wróblewska, Kamila Wojas-Krawczyk, Paweł Krawczyk, Maciej Krzakowski","doi":"10.21037/tlcr-24-552","DOIUrl":"10.21037/tlcr-24-552","url":null,"abstract":"<p><p>Immune checkpoint inhibitors (ICIs) have become an established treatment option for patients with advanced non-small cell lung cancer (NSCLC). However, the efficacy of single-agent immunotherapy as well as in combination with chemotherapy seems to be dependent on the presence of molecular abnormalities in some genes-serine/threonine kinase 11 (<i>STK11</i>), Kelch-like ECH-associated protein 1 (<i>KEAP1</i>) and Kirsten rat sarcoma viral oncogene homolog (<i>KRAS</i>) among them. The <i>KEAP1</i> gene is a critical regulator of the cellular response to oxidative stress and electrophilic stress, thus playing a pivotal role in maintaining cellular homeostasis. The <i>STK11</i> gene encodes a serine/threonine kinase (STK11) involved the regulation of cell growth, polarity, motility, differentiation and cell metabolism. The <i>STK11</i> gene mutations are often associated with an immunologically \"cold\" tumour microenvironment. The co-occurrence of <i>STK11</i> or <i>KEAP1</i> abnormalities with the <i>KRAS</i> mutation changes the composition of the tumour microenvironment as compared when presented alone. The current data, based on retrospective analyses of clinical trials, indicate that the co-existence of <i>STK11</i> and <i>KEAP1</i> genes mutations with the <i>KRAS</i> gene mutations have negative impact on the prognosis, regardless of treatment methods, in patients with advanced NSCLC. However, this group of patients should not be omitted because they constitute a significant percentage of advanced NSCLC patients. Immunotherapy focused on two ICIs [anti-programmed death 1 (PD-1)/anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4)] combined with chemotherapy, may be more effective than immunotherapy or chemotherapy alone in this group of patients. Confirmation of this thesis can be found in the results of available clinical studies. Here, we summarize the theoretical justification as well as the results of clinical trials for combining immunotherapy in patients with <i>STK11</i>-, <i>KEAP1</i>- and <i>KRAS</i>-mutated genes. There is certainly a need to create a prospective clinical trial to assess the effectiveness of combined immunotherapy in the discussed group of patients.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3718-3730"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736579/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tlcr-24-545
Masaya Aoki, Ryo Miyata, Go Kamimura, Shoichiro Morizono, Takuya Tokunaga, Aya Harada-Takeda, Koki Maeda, Toshiyuki Nagata, Kazuhiro Ueda
Background: Perioperative treatment of locally advanced non-small cell lung cancer (NSCLC) is attracting attention. The effect of neoadjuvant tyrosine kinase inhibitor (TKI) therapy on postoperative long-term outcomes in patients with driver gene mutations remains unclear. The aim of this study was to clarify the long-term survival outcomes of patients with stage III NSCLC harboring driver gene mutations who received preoperative TKI therapy.
Methods: Between January 2016 and December 2018, 10 patients with clinical stage III NSCLC with driver gene mutations were treated with TKIs [epidermal growth factor receptor (EGFR) mutation, n=9; anaplastic lymphoma kinase (ALK) fusion, n=1]. One patient refused surgery. The remaining nine patients received sequential chemotherapy followed by surgery. Postoperatively, six patients received adjuvant chemotherapy, and TKIs were readministered in four patients.
Results: The main adverse events of TKIs were grade 3 liver damage and grade 3 skin rash, which required a change in the drug from gefitinib to afatinib and dose reduction, respectively. In all 10 patients, the radiological response to TKIs was greater than the partial response, and nine patients underwent radical surgery. Although viable cancer cells remained in all patients with EGFR mutations, a pathological complete response was obtained in the patient with ALK fusion. No mortality or major morbidity was observed perioperatively. Of the patients who underwent surgery, 3 were alive without recurrence, while 6 had distant metastasis, including 5 with brain metastasis. Seven of the nine patients who underwent surgery were still alive after a median follow-up period of 77.2 months.
Conclusions: Successful long-term outcomes were achieved after sequential targeted therapy and chemotherapy, followed by surgery for stage III NSCLC. However, it is noteworthy that postoperative treatment may have also contributed to minimizing postoperative recurrence.
{"title":"Successful long-term outcome of neoadjuvant sequential targeted therapy and chemotherapy for stage III non-small cell lung carcinoma: 10 case series.","authors":"Masaya Aoki, Ryo Miyata, Go Kamimura, Shoichiro Morizono, Takuya Tokunaga, Aya Harada-Takeda, Koki Maeda, Toshiyuki Nagata, Kazuhiro Ueda","doi":"10.21037/tlcr-24-545","DOIUrl":"10.21037/tlcr-24-545","url":null,"abstract":"<p><strong>Background: </strong>Perioperative treatment of locally advanced non-small cell lung cancer (NSCLC) is attracting attention. The effect of neoadjuvant tyrosine kinase inhibitor (TKI) therapy on postoperative long-term outcomes in patients with driver gene mutations remains unclear. The aim of this study was to clarify the long-term survival outcomes of patients with stage III NSCLC harboring driver gene mutations who received preoperative TKI therapy.</p><p><strong>Methods: </strong>Between January 2016 and December 2018, 10 patients with clinical stage III NSCLC with driver gene mutations were treated with TKIs [epidermal growth factor receptor (<i>EGFR</i>) mutation, n=9; anaplastic lymphoma kinase (<i>ALK</i>) fusion, n=1]. One patient refused surgery. The remaining nine patients received sequential chemotherapy followed by surgery. Postoperatively, six patients received adjuvant chemotherapy, and TKIs were readministered in four patients.</p><p><strong>Results: </strong>The main adverse events of TKIs were grade 3 liver damage and grade 3 skin rash, which required a change in the drug from gefitinib to afatinib and dose reduction, respectively. In all 10 patients, the radiological response to TKIs was greater than the partial response, and nine patients underwent radical surgery. Although viable cancer cells remained in all patients with <i>EGFR</i> mutations, a pathological complete response was obtained in the patient with <i>ALK</i> fusion. No mortality or major morbidity was observed perioperatively. Of the patients who underwent surgery, 3 were alive without recurrence, while 6 had distant metastasis, including 5 with brain metastasis. Seven of the nine patients who underwent surgery were still alive after a median follow-up period of 77.2 months.</p><p><strong>Conclusions: </strong>Successful long-term outcomes were achieved after sequential targeted therapy and chemotherapy, followed by surgery for stage III NSCLC. However, it is noteworthy that postoperative treatment may have also contributed to minimizing postoperative recurrence.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3278-3288"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736588/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012334","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Health-related quality of life (HRQoL) is critical for patients with lung cancer due to poor prognosis. We presented patient-reported outcomes in patients with non-small cell lung cancer (NSCLC) brain metastases (BM) who received whole-brain radiotherapy (WBRT) in combination with erlotinib or WBRT alone in the phase 3 ENTER study.
Methods: The patients' HRQoL was assessed by using the European Organization for Research and Treatment of Cancer 30-item Core Quality of Life Questionnaire (EORTC QLQ-C30). Mean changes in scores on different quality of life (QoL) scales relative to baseline were reported. The QoL response and time to deterioration of QoL were compared between different treatment arms.
Results: The absolute mean differences in global health status/QoL scores, all functional and symptom scale scores from baseline between the two arms were not significantly different at all follow-up time points. After month 5, there was an improvement in nausea/vomiting symptom scores in the WBRT arm relative to their pretreatment baseline (P=0.003), while the WBRT + erlotinib arm had improved scores in fatigue (P=0.01), nausea/vomiting (P=0.02), pain (P=0.04) and insomnia (P=0.01). Compared to the WBRT arm, a greater proportion of patients in the combination treatment arm had deteriorating diarrhea (P=0.009), along with significantly delayed time to deterioration in role function (4.4 vs. 7.0 months, P=0.03), insomnia (7.0 vs. 4.7 months, P=0.02), and constipation (12.7 vs. 9.3 months, P=0.02) symptoms.
Conclusions: The simultaneous addition of erlotinib during WBRT did not decrease the QoL in the overall or epidermal growth factor receptor (EGFR)-mutant patients with BM and resulted in improvements on more QoL scales and slower worsening of some self-reported symptoms compared to WBRT alone.
{"title":"Health-related quality of life analysis from ENTER, a randomized, controlled phase III trial of whole-brain radiotherapy with and without concurrent erlotinib in NSCLC with brain metastases.","authors":"Fei Tang, Jingyi Zhang, Zaicheng Xu, Yu Zhang, Xiaoyue Zhang, Yuan Peng, Zhenzhou Yang","doi":"10.21037/tlcr-24-481","DOIUrl":"10.21037/tlcr-24-481","url":null,"abstract":"<p><strong>Background: </strong>Health-related quality of life (HRQoL) is critical for patients with lung cancer due to poor prognosis. We presented patient-reported outcomes in patients with non-small cell lung cancer (NSCLC) brain metastases (BM) who received whole-brain radiotherapy (WBRT) in combination with erlotinib or WBRT alone in the phase 3 ENTER study.</p><p><strong>Methods: </strong>The patients' HRQoL was assessed by using the European Organization for Research and Treatment of Cancer 30-item Core Quality of Life Questionnaire (EORTC QLQ-C30). Mean changes in scores on different quality of life (QoL) scales relative to baseline were reported. The QoL response and time to deterioration of QoL were compared between different treatment arms.</p><p><strong>Results: </strong>The absolute mean differences in global health status/QoL scores, all functional and symptom scale scores from baseline between the two arms were not significantly different at all follow-up time points. After month 5, there was an improvement in nausea/vomiting symptom scores in the WBRT arm relative to their pretreatment baseline (P=0.003), while the WBRT + erlotinib arm had improved scores in fatigue (P=0.01), nausea/vomiting (P=0.02), pain (P=0.04) and insomnia (P=0.01). Compared to the WBRT arm, a greater proportion of patients in the combination treatment arm had deteriorating diarrhea (P=0.009), along with significantly delayed time to deterioration in role function (4.4 <i>vs.</i> 7.0 months, P=0.03), insomnia (7.0 <i>vs.</i> 4.7 months, P=0.02), and constipation (12.7 <i>vs.</i> 9.3 months, P=0.02) symptoms.</p><p><strong>Conclusions: </strong>The simultaneous addition of erlotinib during WBRT did not decrease the QoL in the overall or epidermal growth factor receptor (EGFR)-mutant patients with BM and resulted in improvements on more QoL scales and slower worsening of some self-reported symptoms compared to WBRT alone.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov identifier: NCT01887795.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"13 12","pages":"3289-3302"},"PeriodicalIF":4.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736605/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012264","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}