Pub Date : 2025-11-30Epub Date: 2025-11-27DOI: 10.21037/tlcr-2025-509
Linda Ye, Ian Dick, Tina Firth, Bruce W Robinson, Jenette Creaney, Alec Redwood
Anti-cancer T cells exhibit a spectrum of functional abilities and exhaustion levels and can be broadly categorized as stem-like exhausted T cells (Texstem) that retain cancer-killing capacity, and terminally exhausted T cells (Texterm) with limited function. Previously, we identified CD8 Texstem and Texterm cells in malignant pleural effusions (PEs) of non-small cell lung cancer (NSCLC) and mesothelioma patients. In both cancers, increased frequency of Texstem cells was associated with improved overall survival (OS). However, not all cancer patients develop PE, and sampling of PE is invasive, with associated risks. In this study we sought to determine if Texstem and Texterm cells in the blood of patients with NSCLC also associated with survival. Using flow cytometry, we quantified Texstem and Texterm in blood samples from 30 patients with advanced NSCLC and assessed their correlation with OS. In half of these cases, we also analyzed matched PE samples for comparison. Compared to PE, peripheral blood had a lower frequency of Texstem (3.8% vs. 9.4% of CD8 T cells, P=0.006) and Texterm (0.3% vs. 1.1%, P=0.002). However, both subsets were significantly correlated between the two sites (Texstem r=0.82, P<0.001; Texterm r=0.63, P=0.01). Higher Texstem cell frequency in the blood was associated with improved survival after adjusting for potentially prognostic patient and tumor related factors. When stratified into high and low groups based on the median, higher Texstem cells levels correlated with better OS [hazard ratio (HR) 0.16, 95% confidence interval (CI): 0.03-0.10, P=0.048]. As shown previously in PE, Texterm cells in the blood did not correlate with OS. Our results further support the importance of Texstem in the anti-cancer immune response and provide useful evidence for the utility of peripheral blood sampling for future studies examining exhausted T cells (Tex).
抗癌T细胞表现出一系列的功能能力和耗竭水平,可以大致分为保留癌症杀伤能力的干细胞样耗竭T细胞(Texstem)和功能有限的终末耗竭T细胞(Texterm)。此前,我们在非小细胞肺癌(NSCLC)和间皮瘤患者的恶性胸腔积液(PEs)中发现了CD8 Texstem和Texterm细胞。在这两种癌症中,Texstem细胞频率的增加与总生存率(OS)的提高有关。然而,并不是所有的癌症患者都会发生PE, PE的采样是侵入性的,有相关的风险。在这项研究中,我们试图确定非小细胞肺癌患者血液中的Texstem和Texterm细胞是否也与生存有关。使用流式细胞术,我们量化了30例晚期NSCLC患者血液样本中的Texstem和Texterm,并评估了它们与OS的相关性。在其中一半的病例中,我们还分析了匹配的PE样本进行比较。与PE相比,外周血中Texstem细胞(3.8% vs. CD8 T细胞9.4%,P=0.006)和Texterm细胞(0.3% vs. 1.1%, P=0.002)的频率较低。然而,这两个亚群在两个位点之间具有显著相关性(Texstem r=0.82, Pterm r=0.63, P=0.01)。在调整潜在预后患者和肿瘤相关因素后,血液中较高的Texstem细胞频率与生存率的提高有关。当根据中位数分为高组和低组时,较高的Texstem细胞水平与较好的OS相关[风险比(HR) 0.16, 95%可信区间(CI): 0.03-0.10, P=0.048]。如先前在PE中所示,血液中的Texterm细胞与OS无关。我们的研究结果进一步支持了Texstem在抗癌免疫反应中的重要性,并为未来研究检测耗尽T细胞(Tex)的外周血取样的实用性提供了有用的证据。
{"title":"Circulating stem-like exhausted CD8 T cells point to better outcomes in lung cancer: a brief report.","authors":"Linda Ye, Ian Dick, Tina Firth, Bruce W Robinson, Jenette Creaney, Alec Redwood","doi":"10.21037/tlcr-2025-509","DOIUrl":"10.21037/tlcr-2025-509","url":null,"abstract":"<p><p>Anti-cancer T cells exhibit a spectrum of functional abilities and exhaustion levels and can be broadly categorized as stem-like exhausted T cells (Tex<sup>stem</sup>) that retain cancer-killing capacity, and terminally exhausted T cells (Tex<sup>term</sup>) with limited function. Previously, we identified CD8 Tex<sup>stem</sup> and Tex<sup>term</sup> cells in malignant pleural effusions (PEs) of non-small cell lung cancer (NSCLC) and mesothelioma patients. In both cancers, increased frequency of Tex<sup>stem</sup> cells was associated with improved overall survival (OS). However, not all cancer patients develop PE, and sampling of PE is invasive, with associated risks. In this study we sought to determine if Tex<sup>stem</sup> and Tex<sup>term</sup> cells in the blood of patients with NSCLC also associated with survival. Using flow cytometry, we quantified Tex<sup>stem</sup> and Tex<sup>term</sup> in blood samples from 30 patients with advanced NSCLC and assessed their correlation with OS. In half of these cases, we also analyzed matched PE samples for comparison. Compared to PE, peripheral blood had a lower frequency of Tex<sup>stem</sup> (3.8% <i>vs.</i> 9.4% of CD8 T cells, P=0.006) and Tex<sup>term</sup> (0.3% <i>vs.</i> 1.1%, P=0.002). However, both subsets were significantly correlated between the two sites (Tex<sup>stem</sup> r=0.82, P<0.001; Tex<sup>term</sup> r=0.63, P=0.01). Higher Tex<sup>stem</sup> cell frequency in the blood was associated with improved survival after adjusting for potentially prognostic patient and tumor related factors. When stratified into high and low groups based on the median, higher Tex<sup>stem</sup> cells levels correlated with better OS [hazard ratio (HR) 0.16, 95% confidence interval (CI): 0.03-0.10, P=0.048]. As shown previously in PE, Tex<sup>term</sup> cells in the blood did not correlate with OS. Our results further support the importance of Tex<sup>stem</sup> in the anti-cancer immune response and provide useful evidence for the utility of peripheral blood sampling for future studies examining exhausted T cells (Tex).</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5074-5081"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683486/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715848","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 : 2025-11-30Epub Date: 2025-11-27DOI: 10.21037/tlcr-2025-422
Birgit Hantzsch-Kuhn, Nina Schraps, Martin Reck, Sönke von Weihe, Till Olchers, David Benjamin Ellebrecht, Katharina Moeller, Christoph Fraune, Maximilian Lennartz, Florian Lutz, Martina Kluth, Georgia Makrypidi-Fraune, Ronald Simon, Guido Sauter, Stefan Steurer
Background: Glucose transporter 1 (GLUT1) is a transmembrane protein responsible for the transportation of glucose across the cell membrane that is often overexpressed in cancer. Due to a key role in cancer glucose metabolism and its membranous localization GLUT1 represents a potential therapeutic target. Our study was designed to elucidate the prevalence of GLUT1 expression and potential associations with tumor phenotype as well as patient outcome in different lung cancer subtypes.
Methods: A tissue microarray containing 858 resected lung cancers was analyzed for GLUT1 expression by immunohistochemistry.
Results: GLUT1 staining was significantly more prevalent and intense in squamous cell carcinoma (SCC, 97.3%) than in pulmonary adenocarcinoma (AC, 62.9%; P<0.001). Of the 225 SCCs, GLUT1 staining was observed in 219 (97.3%) tumors and considered strong in 75.6%, moderate in 15.1%, and weak in 6.7%. In 439 ACs, GLUT1 staining was seen in 276 (62.9%) tumors and considered strong in 14.6%, moderate in 16.4% and weak in 31.9%. High GLUT1 staining was significantly linked to advanced pT stage (P=0.03), nodal metastasis (P<0.001), high grade (P<0.001) and poor overall survival (OS) (P=0.01) in ACs. In SCCs, high GLUT1 staining was unrelated to pT, pN, and histologic grade but significantly linked to OS (P=0.03).
Conclusions: It is concluded that GLUT1 expression is commonly expressed in lung cancer and that a high level of expression is linked to unfavorable tumor features and/or poor prognosis in both AC and SCC.
{"title":"High GLUT1 protein expression is associated with unfavorable tumor features and poor prognosis in non-small cell lung cancer.","authors":"Birgit Hantzsch-Kuhn, Nina Schraps, Martin Reck, Sönke von Weihe, Till Olchers, David Benjamin Ellebrecht, Katharina Moeller, Christoph Fraune, Maximilian Lennartz, Florian Lutz, Martina Kluth, Georgia Makrypidi-Fraune, Ronald Simon, Guido Sauter, Stefan Steurer","doi":"10.21037/tlcr-2025-422","DOIUrl":"10.21037/tlcr-2025-422","url":null,"abstract":"<p><strong>Background: </strong>Glucose transporter 1 (GLUT1) is a transmembrane protein responsible for the transportation of glucose across the cell membrane that is often overexpressed in cancer. Due to a key role in cancer glucose metabolism and its membranous localization GLUT1 represents a potential therapeutic target. Our study was designed to elucidate the prevalence of GLUT1 expression and potential associations with tumor phenotype as well as patient outcome in different lung cancer subtypes.</p><p><strong>Methods: </strong>A tissue microarray containing 858 resected lung cancers was analyzed for GLUT1 expression by immunohistochemistry.</p><p><strong>Results: </strong>GLUT1 staining was significantly more prevalent and intense in squamous cell carcinoma (SCC, 97.3%) than in pulmonary adenocarcinoma (AC, 62.9%; P<0.001). Of the 225 SCCs, GLUT1 staining was observed in 219 (97.3%) tumors and considered strong in 75.6%, moderate in 15.1%, and weak in 6.7%. In 439 ACs, GLUT1 staining was seen in 276 (62.9%) tumors and considered strong in 14.6%, moderate in 16.4% and weak in 31.9%. High GLUT1 staining was significantly linked to advanced pT stage (P=0.03), nodal metastasis (P<0.001), high grade (P<0.001) and poor overall survival (OS) (P=0.01) in ACs. In SCCs, high GLUT1 staining was unrelated to pT, pN, and histologic grade but significantly linked to OS (P=0.03).</p><p><strong>Conclusions: </strong>It is concluded that GLUT1 expression is commonly expressed in lung cancer and that a high level of expression is linked to unfavorable tumor features and/or poor prognosis in both AC and SCC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4838-4848"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683453/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715861","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: Guidelines for driver-negative, resectable stage IIA-IIIB non-small cell lung cancer (NSCLC) recommend surgery combined with neoadjuvant chemotherapy and perioperative immunotherapy. However, the relative efficacy and surgery-related safety comparing various immune checkpoint inhibitors (ICIs) remain unclear. This study aims to directly compare the efficacy and surgery-related safety of perioperative tislelizumab versus pembrolizumab in this setting.
Methods: In this single-institution retrospective cohort study, we enrolled patients with stage IIA-IIIB NSCLC who received neoadjuvant chemotherapy combined with either tislelizumab or pembrolizumab, followed by surgery and adjuvant immunotherapy at our center (2018-2024). Propensity score matching (PSM) was used to balance baseline characteristics. The primary endpoint was pathologic complete response (pCR). Secondary endpoints included major pathologic response (MPR), objective response rate (ORR), overall survival (OS), event-free survival (EFS), and surgery-related safety.
Results: In total, 245 patients were analyzed (75 in the tislelizumab group and 170 in the pembrolizumab group). Following a 1:1 PSM adjustment, each group consisted of 72 patients. No significant differences between tislelizumab group and pembrolizumab group were observed in terms of the pCR rate (43.1% vs. 40.3%; P=0.74), MPR rate (66.8% vs. 66.8%; P>0.99) or ORR (58.3% vs. 66.7%; P=0.30). There were no statistically significant differences between the tislelizumab group and pembrolizumab group in the endpoints of overall OS [hazard ratio (HR) =1.43, 95% confidence interval (CI): 0.59-3.48, P=0.42] and EFS (HR =1.93, 95% CI: 0.96-3.85, P=0.059). There was no significant difference regarding to the surgery-related safety outcomes, including operative time (160.97 vs. 177.80 min, P=0.12), intraoperative blood loss (195.95 vs. 239.73 mL, P=0.28), postoperative chest tube duration (6.594 vs. 7.656 days, P=0.15), and postoperative hospital days (10.00 vs. 10.77 days, P=0.32).
Conclusions: Tislelizumab and pembrolizumab demonstrated comparable drug efficacy and surgery-related safety among patients with resectable stage IIA-IIIB NSCLC undergoing neoadjuvant chemoimmunotherapy.
背景:驱动阴性、可切除的IIA-IIIB期非小细胞肺癌(NSCLC)指南推荐手术联合新辅助化疗和围手术期免疫治疗。然而,比较各种免疫检查点抑制剂(ICIs)的相对疗效和手术相关安全性仍不清楚。本研究旨在直接比较tislelizumab与pembrolizumab在围手术期的疗效和手术相关安全性。方法:在这项单机构回顾性队列研究中,我们招募了IIA-IIIB期NSCLC患者,这些患者接受了新辅助化疗联合替利单抗或派姆单抗,随后进行手术和辅助免疫治疗(2018-2024)。倾向评分匹配(PSM)用于平衡基线特征。主要终点为病理完全缓解(pCR)。次要终点包括主要病理反应(MPR)、客观反应率(ORR)、总生存期(OS)、无事件生存期(EFS)和手术相关安全性。结果:共分析了245例患者(tislelizumab组75例,pembrolizumab组170例)。按1:1调整PSM后,每组72例。tislelizumab组和pembrolizumab组在pCR率(43.1%比40.3%,P=0.74)、MPR率(66.8%比66.8%,P= 0.99)和ORR(58.3%比66.7%,P=0.30)方面无显著差异。tislelizumab组和pembrolizumab组在总OS(风险比(HR) =1.43, 95%可信区间(CI): 0.59-3.48, P=0.42)和EFS (HR =1.93, 95% CI: 0.96-3.85, P=0.059)的终点上无统计学差异。两组手术相关的安全性指标,包括手术时间(160.97 vs. 177.80 min, P=0.12)、术中出血量(195.95 vs. 239.73 mL, P=0.28)、术后胸管持续时间(6.594 vs. 7.656 d, P=0.15)、术后住院天数(10.00 vs. 10.77 d, P=0.32),差异无统计学意义。结论:Tislelizumab和pembrolizumab在接受新辅助化疗免疫治疗的可切除期IIA-IIIB NSCLC患者中显示出相当的药物疗效和手术相关安全性。
{"title":"Comparison of efficacy and surgery-related safety of perioperative tislelizumab and pembrolizumab with neoadjuvant chemotherapy for resectable non-small cell lung cancer: a retrospective cohort study.","authors":"Yizhang Li, Jialing Jiang, Xin Yao, Zhaoyang Wang, Changjian Shao, Jian Wang, Jie Lei, Jinbo Zhao, Masaya Aoki, Hiroyuki Adachi, Yunfeng Ni, Yong Liu, Hongtao Duan, Xiaolong Yan","doi":"10.21037/tlcr-2025-1089","DOIUrl":"10.21037/tlcr-2025-1089","url":null,"abstract":"<p><strong>Background: </strong>Guidelines for driver-negative, resectable stage IIA-IIIB non-small cell lung cancer (NSCLC) recommend surgery combined with neoadjuvant chemotherapy and perioperative immunotherapy. However, the relative efficacy and surgery-related safety comparing various immune checkpoint inhibitors (ICIs) remain unclear. This study aims to directly compare the efficacy and surgery-related safety of perioperative tislelizumab versus pembrolizumab in this setting.</p><p><strong>Methods: </strong>In this single-institution retrospective cohort study, we enrolled patients with stage IIA-IIIB NSCLC who received neoadjuvant chemotherapy combined with either tislelizumab or pembrolizumab, followed by surgery and adjuvant immunotherapy at our center (2018-2024). Propensity score matching (PSM) was used to balance baseline characteristics. The primary endpoint was pathologic complete response (pCR). Secondary endpoints included major pathologic response (MPR), objective response rate (ORR), overall survival (OS), event-free survival (EFS), and surgery-related safety.</p><p><strong>Results: </strong>In total, 245 patients were analyzed (75 in the tislelizumab group and 170 in the pembrolizumab group). Following a 1:1 PSM adjustment, each group consisted of 72 patients. No significant differences between tislelizumab group and pembrolizumab group were observed in terms of the pCR rate (43.1% <i>vs.</i> 40.3%; P=0.74), MPR rate (66.8% <i>vs.</i> 66.8%; P>0.99) or ORR (58.3% <i>vs.</i> 66.7%; P=0.30). There were no statistically significant differences between the tislelizumab group and pembrolizumab group in the endpoints of overall OS [hazard ratio (HR) =1.43, 95% confidence interval (CI): 0.59-3.48, P=0.42] and EFS (HR =1.93, 95% CI: 0.96-3.85, P=0.059). There was no significant difference regarding to the surgery-related safety outcomes, including operative time (160.97 <i>vs.</i> 177.80 min, P=0.12), intraoperative blood loss (195.95 <i>vs.</i> 239.73 mL, P=0.28), postoperative chest tube duration (6.594 <i>vs.</i> 7.656 days, P=0.15), and postoperative hospital days (10.00 <i>vs.</i> 10.77 days, P=0.32).</p><p><strong>Conclusions: </strong>Tislelizumab and pembrolizumab demonstrated comparable drug efficacy and surgery-related safety among patients with resectable stage IIA-IIIB NSCLC undergoing neoadjuvant chemoimmunotherapy.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5017-5030"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683373/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715846","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 and objective: Pulmonary sarcomatoid carcinoma (PSC) accounts for approximately 0.5% of non-small cell lung cancer (NSCLC) cases and is thus a rare subtype. Highly aggressive and hard to detect early, PSC responds poorly to surgery, radiotherapy, and chemotherapy. Therefore, this review aims to synthesize current evidence on its pathogenesis and emerging therapeutic strategies, to improve clinical management.
Methods: We searched PubMed for original studies, reviews, clinical trials, and case reports on PSC published until 2025. Moreover, data from ClinicalTrials.gov and major academic conference proceedings were examined for inclusion in this narrative review.
Key content and findings: The core pathophysiology of PSC is epithelial-mesenchymal transition (EMT), a process that drives biphasic differentiation of tumor cells and remodels the tumor microenvironment (TME), thereby promoting high invasiveness and treatment resistance. Therapeutically, although targetable mutations such as MET exon 14 skipping are relatively frequent in PSC, the efficacy of targeted agents is generally inferior to that for other NSCLC subtypes. Notably, the tumor immune microenvironment of PSC features significant immune cell infiltration and high programmed death-ligand 1 (PD-L1) expression, leading to a generally better response to immune checkpoint inhibitors (ICIs). Consequently, immunotherapy combined with chemotherapy or antiangiogenic agents has emerged as a productive therapeutic strategy.
Conclusions: Precision therapy for PSC, particularly immunotherapy-based combination strategies, has demonstrated transformative potential. However, further efforts in this field should involve clarifying the relevant EMT and tumor heterogeneity mechanisms, optimizing existing treatment regimens, and conducting targeted clinical trials, as these measures may advance individualized precision therapy for patients with PSC and improve their outcomes.
{"title":"The pathogenesis and therapeutic management of rare pulmonary sarcomatoid carcinoma: a narrative review.","authors":"Qianyi Wang, Haoyue Guo, Yuanyuan Wang, Taiping He, Meng Diao, Yuhan Wu, Anwen Xiong, Fei Zhou, Wei Li, Lei Cheng, Chao Zhao, Xuefei Li, Caicun Zhou","doi":"10.21037/tlcr-2025-886","DOIUrl":"10.21037/tlcr-2025-886","url":null,"abstract":"<p><strong>Background and objective: </strong>Pulmonary sarcomatoid carcinoma (PSC) accounts for approximately 0.5% of non-small cell lung cancer (NSCLC) cases and is thus a rare subtype. Highly aggressive and hard to detect early, PSC responds poorly to surgery, radiotherapy, and chemotherapy. Therefore, this review aims to synthesize current evidence on its pathogenesis and emerging therapeutic strategies, to improve clinical management.</p><p><strong>Methods: </strong>We searched PubMed for original studies, reviews, clinical trials, and case reports on PSC published until 2025. Moreover, data from ClinicalTrials.gov and major academic conference proceedings were examined for inclusion in this narrative review.</p><p><strong>Key content and findings: </strong>The core pathophysiology of PSC is epithelial-mesenchymal transition (EMT), a process that drives biphasic differentiation of tumor cells and remodels the tumor microenvironment (TME), thereby promoting high invasiveness and treatment resistance. Therapeutically, although targetable mutations such as <i>MET</i> exon 14 skipping are relatively frequent in PSC, the efficacy of targeted agents is generally inferior to that for other NSCLC subtypes. Notably, the tumor immune microenvironment of PSC features significant immune cell infiltration and high programmed death-ligand 1 (PD-L1) expression, leading to a generally better response to immune checkpoint inhibitors (ICIs). Consequently, immunotherapy combined with chemotherapy or antiangiogenic agents has emerged as a productive therapeutic strategy.</p><p><strong>Conclusions: </strong>Precision therapy for PSC, particularly immunotherapy-based combination strategies, has demonstrated transformative potential. However, further efforts in this field should involve clarifying the relevant EMT and tumor heterogeneity mechanisms, optimizing existing treatment regimens, and conducting targeted clinical trials, as these measures may advance individualized precision therapy for patients with PSC and improve their outcomes.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5137-5158"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683482/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715951","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: Brain metastases (BMs) from non-small cell lung cancer (NSCLC) remain a major clinical challenge, and existing prognostic tools such as the Graded Prognostic Assessment (GPA) do not incorporate imaging biomarkers or adequately reflect the impact of immunotherapy. Meningeal lymphatic vessels (mLVs), which regulate cerebrospinal fluid drainage and immune surveillance, have been implicated in tumor-immune interactions. We aimed to develop and internally validate a multivariable prognostic model integrating mLV remodeling measured by black-blood magnetic resonance imaging (BB-MRI) with clinical predictors to improve early prediction of treatment response.
Methods: We retrospectively analyzed 130 patients with pathologically confirmed NSCLC (100 with BM, 30 without BM). Among the BM cohort, 56 patients achieved favorable treatment response [stable disease (SD) or partial response (PR)] and 44 experienced progressive disease (PD). Candidate predictors were pre-specified based on clinical relevance, and the final model incorporated total mLV diameter, immunotherapy exposure, sex, and extracranial lesion count. Internal validation was performed with 1,000 bootstrap resamples. Model performance was assessed by discrimination, calibration, and decision curve analysis (DCA).
Results: The final multivariable model demonstrated good discrimination [area under the curve (AUC) =0.82; 95% confidence interval (CI): 0.75-0.90], excellent calibration, and consistent net clinical benefit across a range of threshold probabilities. The calibration and decision curves showed promising internal performance, but external validation is required before clinical application.
Conclusions: BB-MRI-derived mLV remodeling may be an early and noninvasive indicator of treatment efficacy in BM. The proposed nomogram enables the individualized prediction of systemic therapy response, supporting precision immunotherapy for patients with intracranial metastases.
{"title":"Development and validation of a multivariable prognostic model incorporating black-blood magnetic resonance imaging-based meningeal lymphatic remodeling to predict therapy response in non-small cell lung cancer brain metastases.","authors":"Wei Shao, Zongbo Li, Feng Qiu, Hengsen Zhang, Shudong Hu, Yifan Liu, Duoduo Li, Yuxi Ge, Hua Lu","doi":"10.21037/tlcr-2025-aw-1154","DOIUrl":"10.21037/tlcr-2025-aw-1154","url":null,"abstract":"<p><strong>Background: </strong>Brain metastases (BMs) from non-small cell lung cancer (NSCLC) remain a major clinical challenge, and existing prognostic tools such as the Graded Prognostic Assessment (GPA) do not incorporate imaging biomarkers or adequately reflect the impact of immunotherapy. Meningeal lymphatic vessels (mLVs), which regulate cerebrospinal fluid drainage and immune surveillance, have been implicated in tumor-immune interactions. We aimed to develop and internally validate a multivariable prognostic model integrating mLV remodeling measured by black-blood magnetic resonance imaging (BB-MRI) with clinical predictors to improve early prediction of treatment response.</p><p><strong>Methods: </strong>We retrospectively analyzed 130 patients with pathologically confirmed NSCLC (100 with BM, 30 without BM). Among the BM cohort, 56 patients achieved favorable treatment response [stable disease (SD) or partial response (PR)] and 44 experienced progressive disease (PD). Candidate predictors were pre-specified based on clinical relevance, and the final model incorporated total mLV diameter, immunotherapy exposure, sex, and extracranial lesion count. Internal validation was performed with 1,000 bootstrap resamples. Model performance was assessed by discrimination, calibration, and decision curve analysis (DCA).</p><p><strong>Results: </strong>The final multivariable model demonstrated good discrimination [area under the curve (AUC) =0.82; 95% confidence interval (CI): 0.75-0.90], excellent calibration, and consistent net clinical benefit across a range of threshold probabilities. The calibration and decision curves showed promising internal performance, but external validation is required before clinical application.</p><p><strong>Conclusions: </strong>BB-MRI-derived mLV remodeling may be an early and noninvasive indicator of treatment efficacy in BM. The proposed nomogram enables the individualized prediction of systemic therapy response, supporting precision immunotherapy for patients with intracranial metastases.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5059-5073"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683432/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715789","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: SMARCA4-deficient non-small cell lung cancer (SD-NSCLC) is a highly aggressive malignancy with a poor prognosis, and the clinical efficacy of immune checkpoint inhibitors (ICIs) in this context remains under investigation. This study focuses on patients with SD-NSCLC, investigating the efficacy of ICIs and the risk factors affecting prognosis, while also conducting a preliminary exploration of the underlying mechanisms through The Cancer Genome Atlas (TCGA) database.
Methods: From October 2020 to May 2025, 95 patients with SD-NSCLC, confirmed by immunohistochemistry (IHC) at Tianjin Medical University Cancer Institute & Hospital, were included for analysis. Validation and molecular mechanism exploration were conducted using the TCGA database. Disease-free survival (DFS) and progression-free survival (PFS) were the primary endpoints of the study. The disease control rate (DCR) was a secondary endpoint.
Results: In stage IV SD-NSCLC patients, no significant difference in PFS was observed between those treated with ICIs and non-ICIs (P=0.60). However, the median PFS (mPFS) differences were significant between STK11/KEAP1 mutant-type (mut) and wild-type (wt) groups (1.0 vs. 6.5 months, P=0.007). Even after ICI treatment, the difference in mPFS between the STK11/KEAP1 wt and mut groups remained significant (P=0.02). Additionally, there was a significant difference in the mPFS between the SD group treated with ICIs and the non-SD group (6.0 vs. 11.0 months, P=0.003). TCGA analysis revealed that SMARCA4 loss-of-function (LOF) mutations had significantly shorter mPFS compared to non-LOF mutations (18.66 vs. 57.56 months, P=0.02). Differential gene expression and enrichment analysis, along with immune infiltration analysis, revealed that SMARCA4-LOF was associated with gene silencing and immune suppression, which may explain the limited efficacy of ICIs.
Conclusions: SMARCA4 deficiency is an independent prognostic factor for NSCLC, as validated using the TCGA database. Co-mutations with STK11/KEAP1 further exacerbate poor prognosis, suggesting that specific gene co-mutations may influence treatment response and survival. Moreover, SMARCA4 deficiency leads to poor responses to immunotherapy, potentially due to core metabolic disorders, inactivation of tumor suppressor signaling, and immune suppression in the tumor microenvironment. These findings suggest that treatment strategies should be adjusted to address these molecular mechanisms.
背景:smarca4缺陷的非小细胞肺癌(SD-NSCLC)是一种预后不良的高侵袭性恶性肿瘤,免疫检查点抑制剂(ICIs)在这种情况下的临床疗效仍在研究中。本研究以SD-NSCLC患者为研究对象,探讨ICIs的疗效及影响预后的危险因素,同时通过the Cancer Genome Atlas (TCGA)数据库对其潜在机制进行初步探讨。方法:选取2020年10月至2025年5月天津医科大学肿瘤研究所附属医院经免疫组化(IHC)确诊的SD-NSCLC患者95例进行分析。利用TCGA数据库进行验证和分子机制探索。无病生存期(DFS)和无进展生存期(PFS)是研究的主要终点。疾病控制率(DCR)是次要终点。结果:在IV期SD-NSCLC患者中,使用ICIs与未使用ICIs治疗的PFS无显著差异(P=0.60)。然而,STK11/KEAP1突变型(mut)组和野生型(wt)组的中位PFS (mPFS)差异显著(1.0个月vs. 6.5个月,P=0.007)。即使在ICI治疗后,STK11/KEAP1 wt组和mut组之间的mPFS差异仍然显著(P=0.02)。此外,接受ICIs治疗的SD组与非SD组的mPFS有显著差异(6.0个月vs 11.0个月,P=0.003)。TCGA分析显示,与非LOF突变相比,SMARCA4功能丧失(LOF)突变的mPFS显著缩短(18.66个月对57.56个月,P=0.02)。差异基因表达和富集分析以及免疫浸润分析显示,SMARCA4-LOF与基因沉默和免疫抑制有关,这可能解释了ICIs疗效有限的原因。结论:SMARCA4缺乏是NSCLC的独立预后因素,TCGA数据库证实了这一点。与STK11/KEAP1共突变进一步加剧了不良预后,表明特异性基因共突变可能影响治疗反应和生存。此外,SMARCA4缺乏导致对免疫治疗的不良反应,可能是由于核心代谢紊乱、肿瘤抑制信号失活和肿瘤微环境中的免疫抑制。这些发现表明,治疗策略应该调整,以解决这些分子机制。
{"title":"Immune checkpoint inhibitors have limited efficacy in <i>SMARCA4</i>-deficient non-small cell lung cancer.","authors":"Ying Han, Jing Wang, Boyue Pang, Jiali Zhang, Xiaoliang Zhao, Shuan Hao, Qiang Zhang, Xiubao Ren, Leina Sun","doi":"10.21037/tlcr-2025-921","DOIUrl":"10.21037/tlcr-2025-921","url":null,"abstract":"<p><strong>Background: </strong><i>SMARCA4</i>-deficient non-small cell lung cancer (SD-NSCLC) is a highly aggressive malignancy with a poor prognosis, and the clinical efficacy of immune checkpoint inhibitors (ICIs) in this context remains under investigation. This study focuses on patients with SD-NSCLC, investigating the efficacy of ICIs and the risk factors affecting prognosis, while also conducting a preliminary exploration of the underlying mechanisms through The Cancer Genome Atlas (TCGA) database.</p><p><strong>Methods: </strong>From October 2020 to May 2025, 95 patients with SD-NSCLC, confirmed by immunohistochemistry (IHC) at Tianjin Medical University Cancer Institute & Hospital, were included for analysis. Validation and molecular mechanism exploration were conducted using the TCGA database. Disease-free survival (DFS) and progression-free survival (PFS) were the primary endpoints of the study. The disease control rate (DCR) was a secondary endpoint.</p><p><strong>Results: </strong>In stage IV SD-NSCLC patients, no significant difference in PFS was observed between those treated with ICIs and non-ICIs (P=0.60). However, the median PFS (mPFS) differences were significant between <i>STK11</i>/<i>KEAP1</i> mutant-type (mut) and wild-type (wt) groups (1.0 <i>vs.</i> 6.5 months, P=0.007). Even after ICI treatment, the difference in mPFS between the <i>STK11</i>/<i>KEAP1</i> wt and mut groups remained significant (P=0.02). Additionally, there was a significant difference in the mPFS between the SD group treated with ICIs and the non-SD group (6.0 <i>vs.</i> 11.0 months, P=0.003). TCGA analysis revealed that <i>SMARCA4</i> loss-of-function (LOF) mutations had significantly shorter mPFS compared to non-LOF mutations (18.66 <i>vs.</i> 57.56 months, P=0.02). Differential gene expression and enrichment analysis, along with immune infiltration analysis, revealed that <i>SMARCA4</i>-LOF was associated with gene silencing and immune suppression, which may explain the limited efficacy of ICIs.</p><p><strong>Conclusions: </strong><i>SMARCA4</i> deficiency is an independent prognostic factor for NSCLC, as validated using the TCGA database. Co-mutations with <i>STK11/KEAP1</i> further exacerbate poor prognosis, suggesting that specific gene co-mutations may influence treatment response and survival. Moreover, <i>SMARCA4</i> deficiency leads to poor responses to immunotherapy, potentially due to core metabolic disorders, inactivation of tumor suppressor signaling, and immune suppression in the tumor microenvironment. These findings suggest that treatment strategies should be adjusted to address these molecular mechanisms.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5000-5016"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715839","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 : 2025-11-30Epub Date: 2025-11-27DOI: 10.21037/tlcr-2025-1049
Yuchen Huang, Jian Zhou, Cheng Shen, Ruijia Yang, Quan Zheng, Yang Qiu, Xiaoli Mei, Kunhao Wu, Dongsheng Wu, Jinghan Wang, Wenjing Jiang, Hu Liao
Background: Anaplastic lymphoma kinase (ALK) rearrangement is a key driver mutation in lung adenocarcinoma (LUAD). Despite its established role in advanced diseases, the prognostic impact of ALK rearrangement on early-stage LUAD remains unexplored. Therefore, we aimed to explore the prognostic value of ALK rearrangement in surgically treated stage I LUAD by analyzing patient demographics, tumor characteristics, recurrence, and therapy patterns.
Methods: This retrospective study reviewed postoperative pathologic stage I LUAD patients who underwent ALK rearrangement testing at West China Hospital, Sichuan University. The prognostic impact of ALK rearrangement on recurrence-free survival (RFS), along with clinicopathologic features, mutational profiles, and targeted therapy patterns of ALK-positive patients, was assessed.
Results: The cohort comprised 3,775 patients, of whom 165 (4.37%) were ALK-positive. ALK rearrangement rates were significantly higher in females (P=0.048), stage IB (P<0.001), and those without epidermal growth factor receptor (EGFR) mutations (P<0.001). ALK-positive tumors were more likely to exhibit high-risk radio-pathological features for recurrence, with the trend being more pronounced in stage IA cases. ALK-positive patients showed worse RFS compared to negative ones (log-rank P=0.009), but this difference did not remain significant in multivariable analysis [hazard ratio (HR) =1.72; P=0.09]. Further analysis revealed that ALK rearrangement was significantly associated with an increased risk of metastasis [odds ratio (OR) =2.15; P=0.045], particularly to the bone (OR =5.38; P<0.001) and brain (OR =2.88; P=0.041) after adjusting for confounders.
Conclusions: ALK rearrangements were linked to more aggressive histology and increased risk of bone and brain metastases in resected, stage I LUADs, emphasizing the potential relevance of molecular profiling in postoperative risk assessment and treatment planning, even in stage IA patients.
{"title":"Prognostic impact of <i>ALK</i> rearrangement in surgically treated stage I lung adenocarcinoma: a retrospective cohort study.","authors":"Yuchen Huang, Jian Zhou, Cheng Shen, Ruijia Yang, Quan Zheng, Yang Qiu, Xiaoli Mei, Kunhao Wu, Dongsheng Wu, Jinghan Wang, Wenjing Jiang, Hu Liao","doi":"10.21037/tlcr-2025-1049","DOIUrl":"10.21037/tlcr-2025-1049","url":null,"abstract":"<p><strong>Background: </strong>Anaplastic lymphoma kinase (<i>ALK</i>) rearrangement is a key driver mutation in lung adenocarcinoma (LUAD). Despite its established role in advanced diseases, the prognostic impact of <i>ALK</i> rearrangement on early-stage LUAD remains unexplored. Therefore, we aimed to explore the prognostic value of <i>ALK</i> rearrangement in surgically treated stage I LUAD by analyzing patient demographics, tumor characteristics, recurrence, and therapy patterns.</p><p><strong>Methods: </strong>This retrospective study reviewed postoperative pathologic stage I LUAD patients who underwent <i>ALK</i> rearrangement testing at West China Hospital, Sichuan University. The prognostic impact of <i>ALK</i> rearrangement on recurrence-free survival (RFS), along with clinicopathologic features, mutational profiles, and targeted therapy patterns of <i>ALK</i>-positive patients, was assessed.</p><p><strong>Results: </strong>The cohort comprised 3,775 patients, of whom 165 (4.37%) were <i>ALK</i>-positive. <i>ALK</i> rearrangement rates were significantly higher in females (P=0.048), stage IB (P<0.001), and those without epidermal growth factor receptor (<i>EGFR</i>) mutations (P<0.001). <i>ALK</i>-positive tumors were more likely to exhibit high-risk radio-pathological features for recurrence, with the trend being more pronounced in stage IA cases. <i>ALK</i>-positive patients showed worse RFS compared to negative ones (log-rank P=0.009), but this difference did not remain significant in multivariable analysis [hazard ratio (HR) =1.72; P=0.09]. Further analysis revealed that <i>ALK</i> rearrangement was significantly associated with an increased risk of metastasis [odds ratio (OR) =2.15; P=0.045], particularly to the bone (OR =5.38; P<0.001) and brain (OR =2.88; P=0.041) after adjusting for confounders.</p><p><strong>Conclusions: </strong><i>ALK</i> rearrangements were linked to more aggressive histology and increased risk of bone and brain metastases in resected, stage I LUADs, emphasizing the potential relevance of molecular profiling in postoperative risk assessment and treatment planning, even in stage IA patients.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4824-4837"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12685409/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715798","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 and objective: The increasing use of neoadjuvant therapy in non-small cell lung cancer (NSCLC) has magnified the importance of pathologic response as a treatment endpoint. However, there are persistent challenges in its assessment and interpretation. This review aimed to synthesize current methods and challenges in evaluating pathologic response in patients with NSCLC, summarize available assessment techniques and biomarkers, and collate current data on pathologic response to neoadjuvant treatments and the association with survival outcomes.
Methods: We selected and reviewed articles proposing guidelines or approaches to the pathologic assessment of NSCLC, articles describing challenges in assessing pathologic response in NSCLC, and studies reporting pathologic response to neoadjuvant treatment and/or the association between pathologic response and survival outcomes. Data were extracted and summarized descriptively.
Key content and findings: In this review, we summarize methods for evaluating pathologic response in patients with resectable NSCLC and highlight current challenges, including variability in pathologic response assessment, the limited standardization of techniques and biomarkers, and the difficulty of interpreting pathologic response. We also review current clinical data on pathologic response to neoadjuvant chemotherapy, radiotherapy, immunotherapy, tyrosine kinase inhibitors (TKIs), and antiangiogenic therapies, and the association between pathologic response and survival outcomes. Finally, we review and discuss the selection of optimal treatment strategies for patients who achieve a pathologic response to neoadjuvant therapy.
Conclusions: Pathologic response is a valuable indicator of early response to treatment, but its current limitations necessitate a cautious and balanced approach in treatment decision-making for patients with early-stage resectable NSCLC, with consideration also given to other factors such as long-term survival and quality of life.
{"title":"A narrative review of pathologic response in non-small cell lung cancer: challenges, implications, and future directions.","authors":"Yuan Li, Fei Liang, Suijun Xiao, Chao Yuan, Haiquan Chen","doi":"10.21037/tlcr-2025-584","DOIUrl":"10.21037/tlcr-2025-584","url":null,"abstract":"<p><strong>Background and objective: </strong>The increasing use of neoadjuvant therapy in non-small cell lung cancer (NSCLC) has magnified the importance of pathologic response as a treatment endpoint. However, there are persistent challenges in its assessment and interpretation. This review aimed to synthesize current methods and challenges in evaluating pathologic response in patients with NSCLC, summarize available assessment techniques and biomarkers, and collate current data on pathologic response to neoadjuvant treatments and the association with survival outcomes.</p><p><strong>Methods: </strong>We selected and reviewed articles proposing guidelines or approaches to the pathologic assessment of NSCLC, articles describing challenges in assessing pathologic response in NSCLC, and studies reporting pathologic response to neoadjuvant treatment and/or the association between pathologic response and survival outcomes. Data were extracted and summarized descriptively.</p><p><strong>Key content and findings: </strong>In this review, we summarize methods for evaluating pathologic response in patients with resectable NSCLC and highlight current challenges, including variability in pathologic response assessment, the limited standardization of techniques and biomarkers, and the difficulty of interpreting pathologic response. We also review current clinical data on pathologic response to neoadjuvant chemotherapy, radiotherapy, immunotherapy, tyrosine kinase inhibitors (TKIs), and antiangiogenic therapies, and the association between pathologic response and survival outcomes. Finally, we review and discuss the selection of optimal treatment strategies for patients who achieve a pathologic response to neoadjuvant therapy.</p><p><strong>Conclusions: </strong>Pathologic response is a valuable indicator of early response to treatment, but its current limitations necessitate a cautious and balanced approach in treatment decision-making for patients with early-stage resectable NSCLC, with consideration also given to other factors such as long-term survival and quality of life.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5118-5136"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683421/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715818","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 : 2025-11-30Epub Date: 2025-11-20DOI: 10.21037/tlcr-2025-883
Xavier Fremand, Ilaria Onorati, Boris Duchemann
{"title":"Perioperative chemo immunotherapy in non-small-cell lung cancer: what can we learn from the long-term results of NADIM?","authors":"Xavier Fremand, Ilaria Onorati, Boris Duchemann","doi":"10.21037/tlcr-2025-883","DOIUrl":"10.21037/tlcr-2025-883","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4704-4710"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683400/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715862","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 mesenchymal-epithelial transition factor (MET) gene is a key therapeutic target in non-small cell lung cancer (NSCLC), affecting the tumor expression of programmed cell death ligand 1 (PD-L1) and the immune microenvironment. This study examined the relationship between cellular MET (c-MET) expression, immune profiles, and survival in patients with NSCLC treated with radiochemotherapy and consolidative immunotherapy.
Methods: c-MET expression levels and immune profiles, including cluster of differentiation 8 (CD8), cluster of differentiation 68 (CD68), lymphocyte-activation gene 3 (LAG3), forkhead box protein P3 (FOXP3), and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), were accessed in 60 locally advanced patients with NSCLC treated with radiochemotherapy and sequential immunotherapy at Shanghai Pulmonary Hospital. Correlations between c-MET gene expression and immune cell infiltration were additionally explored using publicly available transcriptomic datasets and bioinformatics analysis. Categorical data were analyzed with Chi-squared tests, survival functions were derived from Kaplan-Meier estimates, and Cox regression assessed the independent impact on overall survival (OS) and progression-free survival (PFS).
Results: PFS and OS for the cohort were 24.0 and 34.0 months, respectively. OS was significantly associated with reduced macrophage (CD68+) infiltration (P=0.03), while PFS was significantly associated with high c-MET expression (P=0.03). Correlation analysis indicated a significant correlation between c-MET expression and macrophage (CD68+) infiltration (P=0.01).
Conclusions: c-MET expression and macrophage (CD68+) infiltration were identified as potential predictors of survival and were found to be significantly correlated with one another. These findings suggest that antibody-drug conjugates targeting c-MET in combination with immune checkpoint inhibitors may represent a potentially effective consolidation therapy strategy following chemoradiotherapy in patients with locally advanced NSCLC.
{"title":"c-MET expression and immune landscape in locally advanced patients with non-small cell lung cancer undergoing radiochemotherapy and consolidative immunotherapy.","authors":"Lisha Ye, Xiaoling Xu, Ying Zhang, Wei Zhang, Leilei Wu, Chunyan Wu, Yaoyao Zhu, Yaping Xu","doi":"10.21037/tlcr-2025-1036","DOIUrl":"10.21037/tlcr-2025-1036","url":null,"abstract":"<p><strong>Background: </strong>The mesenchymal-epithelial transition factor (<i>MET</i>) gene is a key therapeutic target in non-small cell lung cancer (NSCLC), affecting the tumor expression of programmed cell death ligand 1 (PD-L1) and the immune microenvironment. This study examined the relationship between cellular MET (c-MET) expression, immune profiles, and survival in patients with NSCLC treated with radiochemotherapy and consolidative immunotherapy.</p><p><strong>Methods: </strong>c-MET expression levels and immune profiles, including cluster of differentiation 8 (CD8), cluster of differentiation 68 (CD68), lymphocyte-activation gene 3 (<i>LAG3</i>), forkhead box protein P3 (FOXP3), and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), were accessed in 60 locally advanced patients with NSCLC treated with radiochemotherapy and sequential immunotherapy at Shanghai Pulmonary Hospital. Correlations between <i>c-MET</i> gene expression and immune cell infiltration were additionally explored using publicly available transcriptomic datasets and bioinformatics analysis. Categorical data were analyzed with Chi-squared tests, survival functions were derived from Kaplan-Meier estimates, and Cox regression assessed the independent impact on overall survival (OS) and progression-free survival (PFS).</p><p><strong>Results: </strong>PFS and OS for the cohort were 24.0 and 34.0 months, respectively. OS was significantly associated with reduced macrophage (CD68<sup>+</sup>) infiltration (P=0.03), while PFS was significantly associated with high c-MET expression (P=0.03). Correlation analysis indicated a significant correlation between c-MET expression and macrophage (CD68<sup>+</sup>) infiltration (P=0.01).</p><p><strong>Conclusions: </strong>c-MET expression and macrophage (CD68<sup>+</sup>) infiltration were identified as potential predictors of survival and were found to be significantly correlated with one another. These findings suggest that antibody-drug conjugates targeting c-MET in combination with immune checkpoint inhibitors may represent a potentially effective consolidation therapy strategy following chemoradiotherapy in patients with locally advanced NSCLC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5031-5043"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715871","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}