Pub Date : 2025-10-31Epub Date: 2025-10-29DOI: 10.21037/tlcr-2025-642
Marc G Denis
{"title":"Molecular minimal residual disease in resected non-small cell lung cancer: \"flattening the curve\".","authors":"Marc G Denis","doi":"10.21037/tlcr-2025-642","DOIUrl":"10.21037/tlcr-2025-642","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4180-4183"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605196/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514020","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: With advances in multimodal treatment, the importance of identifying biomarkers by bronchoscopy has increased, even in potentially resectable non-small cell lung cancer (NSCLC). However, it remains unclear whether next-generation sequencing (NGS) using bronchoscopic specimens is as useful in potentially resectable NSCLC as in unresectable cases. The objective of this study was to evaluate the feasibility of performing NGS using bronchoscopic specimens obtained from patients with potentially resectable NSCLC.
Methods: The patients diagnosed with NSCLC via bronchoscopy at National Cancer Center Hospital who underwent NGS were retrospectively analyzed. We allocated all N0-2 and M0 and N3 and/or M1 cases to the potentially resectable and unresectable groups, respectively. Patients were divided into three subgroups according to the target: endobronchial, peri-tracheal/bronchial, and lung lesions. The NGS analytical success rates for potentially resectable NSCLC were compared with unresectable cases for each subgroup to investigate the feasibility of NGS-targeted bronchoscopy.
Results: Among the 644 patients, 184 had potentially resectable disease, and 460 had unresectable disease. The overall NGS analytical success rate was significantly lower in the potentially resectable group than in unresectable group (81.0% vs. 87.8%, P=0.03). Concerning endobronchial and peri-tracheal/bronchial lesions, there were no significant differences in the NGS analytical success rates (85.7% vs. 100%, P=0.48; and 84.9% vs. 88.2%, P=0.44). The NGS analytical success rate for lung lesions was significantly lower in the potentially resectable group (73.4% vs. 86.4%, P=0.03). Inner-located tumors (P=0.04) were significant factors associated with analytical success.
Conclusions: The overall rate of successful NGS analysis using bronchoscopic specimens were lower in potentially resectable NSCLC cases, especially in lung lesions. To improve NGS feasibility, it is important to adequately harvest lung lesions and prioritize targeting endobronchial or peri-tracheal/bronchial lesions.
背景:随着多模式治疗的进展,通过支气管镜识别生物标志物的重要性增加,即使在可能可切除的非小细胞肺癌(NSCLC)中也是如此。然而,尚不清楚使用支气管镜标本的下一代测序(NGS)在可能可切除的非小细胞肺癌中是否与不可切除的病例一样有用。本研究的目的是评估使用可能可切除的非小细胞肺癌患者的支气管镜标本进行NGS的可行性。方法:回顾性分析国立肿瘤中心医院经支气管镜诊断为非小细胞肺癌并行NGS的患者。我们将所有N0-2和M0以及N3和/或M1病例分别分配到潜在可切除组和不可切除组。根据目标将患者分为三个亚组:支气管内、气管周围/支气管和肺部病变。将每个亚组中可切除NSCLC的NGS分析成功率与不可切除病例进行比较,以探讨NGS靶向支气管镜检查的可行性。结果:644例患者中,可切除病变184例,不可切除病变460例。可切除组总体NGS分析成功率明显低于不可切除组(81.0% vs 87.8%, P=0.03)。对于支气管内和气管周围/支气管病变,NGS分析成功率无显著差异(85.7% vs 100%, P=0.48; 84.9% vs 88.2%, P=0.44)。可切除组肺病变的NGS分析成功率明显较低(73.4%比86.4%,P=0.03)。内部肿瘤(P=0.04)是分析成功的重要因素。结论:在潜在可切除的非小细胞肺癌病例中,特别是肺病变病例中,支气管镜标本NGS分析的总体成功率较低。为了提高NGS的可行性,重要的是要充分收集肺部病变,优先针对支气管内或气管周围/支气管病变。
{"title":"Feasibility of next-generation sequencing using bronchoscopic specimens in potentially resectable non-small cell lung cancer.","authors":"Kaito Yano, Yuji Matsumoto, Hideaki Furuse, Keigo Uchimura, Tatsuya Imabayashi, Keisuke Asakura, Takaaki Tsuchida","doi":"10.21037/tlcr-2025-605","DOIUrl":"10.21037/tlcr-2025-605","url":null,"abstract":"<p><strong>Background: </strong>With advances in multimodal treatment, the importance of identifying biomarkers by bronchoscopy has increased, even in potentially resectable non-small cell lung cancer (NSCLC). However, it remains unclear whether next-generation sequencing (NGS) using bronchoscopic specimens is as useful in potentially resectable NSCLC as in unresectable cases. The objective of this study was to evaluate the feasibility of performing NGS using bronchoscopic specimens obtained from patients with potentially resectable NSCLC.</p><p><strong>Methods: </strong>The patients diagnosed with NSCLC via bronchoscopy at National Cancer Center Hospital who underwent NGS were retrospectively analyzed. We allocated all N0-2 and M0 and N3 and/or M1 cases to the potentially resectable and unresectable groups, respectively. Patients were divided into three subgroups according to the target: endobronchial, peri-tracheal/bronchial, and lung lesions. The NGS analytical success rates for potentially resectable NSCLC were compared with unresectable cases for each subgroup to investigate the feasibility of NGS-targeted bronchoscopy.</p><p><strong>Results: </strong>Among the 644 patients, 184 had potentially resectable disease, and 460 had unresectable disease. The overall NGS analytical success rate was significantly lower in the potentially resectable group than in unresectable group (81.0% <i>vs.</i> 87.8%, P<i>=</i>0.03). Concerning endobronchial and peri-tracheal/bronchial lesions, there were no significant differences in the NGS analytical success rates (85.7% <i>vs.</i> 100%, P<i>=</i>0.48; and 84.9% <i>vs.</i> 88.2%, P<i>=</i>0.44). The NGS analytical success rate for lung lesions was significantly lower in the potentially resectable group (73.4% <i>vs.</i> 86.4%, P<i>=</i>0.03). Inner-located tumors (P<i>=</i>0.04) were significant factors associated with analytical success.</p><p><strong>Conclusions: </strong>The overall rate of successful NGS analysis using bronchoscopic specimens were lower in potentially resectable NSCLC cases, especially in lung lesions. To improve NGS feasibility, it is important to adequately harvest lung lesions and prioritize targeting endobronchial or peri-tracheal/bronchial lesions.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4268-4284"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605578/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514248","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 current standard of care for unresectable stage III non-small cell lung cancer (NSCLC) is concurrent chemoradiotherapy (CCRT) followed by durvalumab. There are no reliable biomarkers predicting the response to this treatment. We aimed to analyze the relationship between soluble immune mediators and treatment response and to identify biomarkers predicting the treatment response in patients with unresectable stage III NSCLC.
Methods: We prospectively enrolled 48 patients diagnosed with stage III NSCLC who underwent thoracic radiotherapy (cohort A). Of these patients, 29 received durvalumab after CCRT (cohort B). Peripheral blood samples were collected before radiotherapy and 6 weeks after durvalumab treatment. To identify soluble immune mediators associated with progression-free survival (PFS) and overall survival (OS), we examined the levels of 83 soluble immune mediators using a bead-based multiplex assay and calculated the concordance index (C-index).
Results: At baseline, the high matrix metalloproteinase (MMP)-2 group exhibited the longest PFS and OS in cohort A (C-index: 0.70 and 0.75, respectively), whereas the high IFN-γ group had the worst PFS and OS in cohort B (C-index: 0.75 and 0.71, respectively). Furthermore, patients with greater increases in CXCL9, CXCL10, and CCL23 levels after durvalumab treatment had poor PFS and OS in cohort B (C-index >0.70).
Conclusions: Our results suggest that PFS and OS are negatively affected by elevated CXCL9, CXCL10, and CCL23 levels after CCRT followed by durvalumab treatment. These soluble mediators may be useful tools for the clinical management of locally advanced NSCLC treated with PD-L1 therapy after CCRT.
{"title":"Association between soluble immune mediators and outcomes in patients with unresectable stage III non-small cell lung cancer treated with concurrent chemoradiotherapy followed by durvalumab.","authors":"Takaaki Tokito, Koichi Azuma, Kenta Murotani, Norikazu Matsuo, Goushi Matama, Daiki Murata, Tetsuro Sasada, Tomoaki Hoshino","doi":"10.21037/tlcr-2025-563","DOIUrl":"10.21037/tlcr-2025-563","url":null,"abstract":"<p><strong>Background: </strong>The current standard of care for unresectable stage III non-small cell lung cancer (NSCLC) is concurrent chemoradiotherapy (CCRT) followed by durvalumab. There are no reliable biomarkers predicting the response to this treatment. We aimed to analyze the relationship between soluble immune mediators and treatment response and to identify biomarkers predicting the treatment response in patients with unresectable stage III NSCLC.</p><p><strong>Methods: </strong>We prospectively enrolled 48 patients diagnosed with stage III NSCLC who underwent thoracic radiotherapy (cohort A). Of these patients, 29 received durvalumab after CCRT (cohort B). Peripheral blood samples were collected before radiotherapy and 6 weeks after durvalumab treatment. To identify soluble immune mediators associated with progression-free survival (PFS) and overall survival (OS), we examined the levels of 83 soluble immune mediators using a bead-based multiplex assay and calculated the concordance index (C-index).</p><p><strong>Results: </strong>At baseline, the high matrix metalloproteinase (MMP)-2 group exhibited the longest PFS and OS in cohort A (C-index: 0.70 and 0.75, respectively), whereas the high IFN-γ group had the worst PFS and OS in cohort B (C-index: 0.75 and 0.71, respectively). Furthermore, patients with greater increases in CXCL9, CXCL10, and CCL23 levels after durvalumab treatment had poor PFS and OS in cohort B (C-index >0.70).</p><p><strong>Conclusions: </strong>Our results suggest that PFS and OS are negatively affected by elevated CXCL9, CXCL10, and CCL23 levels after CCRT followed by durvalumab treatment. These soluble mediators may be useful tools for the clinical management of locally advanced NSCLC treated with PD-L1 therapy after CCRT.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4449-4459"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605586/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514084","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: Combination immunotherapy has become a standard first-line treatment for advanced non-small cell lung cancer (NSCLC). However, its application in patients with checkpoint inhibitor-related pneumonitis (CIP) remains challenging due to the risk of treatment-related pulmonary toxicity. While first-line outcomes in CIP patients are increasingly reported, data on the efficacy and safety of subsequent therapies are lacking. This study aimed to evaluate treatment patterns, survival outcomes, and the risk of pulmonary toxicity associated with second-line therapy in NSCLC patients with prior CIP after first-line immunotherapy.
Methods: This retrospective study analyzed 159 patients with advanced or recurrent NSCLC treated with first-line combination immunotherapy at Fukuoka University Hospital between January 2019 and March 2024, including those who had previously received molecular targeted therapy for driver gene mutations. Patients were stratified based on baseline CIP status. Among them, 91 patients (19 prior CIP and 72 non-prior CIP) who received second-line therapy were evaluated for progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and drug-induced pneumonitis onset during second-line treatment.
Results: Baseline CIP was present in 32 patients (20.1%). The transition rate to second-line therapy was 73% in the prior CIP group and 64% in the non-prior CIP group, with no statistically significant difference observed between the two groups (P=0.49), indicating that the presence of prior CIP did not clearly affect the likelihood of receiving secondary therapy. PFS following second-line treatment was similar between patients with and without prior CIP. However, OS tended to be shorter in the prior CIP group (6.4 vs. 10.3 months), although the difference was not statistically significant (P=0.07), a trend that persisted after propensity score matching. Drug-induced pneumonitis development during second-line therapy occurred more frequently in patients with prior CIP (26% vs. 3%, P<0.01).
Conclusions: Second-line cytotoxic chemotherapy following combination immunotherapy may be associated with shorter survival and increased pulmonary toxicity in NSCLC patients with prior CIP. Careful clinical decision-making, multidisciplinary consultation, and close monitoring are essential when initiating second-line treatment in this population.
背景:联合免疫治疗已成为晚期非小细胞肺癌(NSCLC)的标准一线治疗方法。然而,由于存在治疗相关肺毒性的风险,其在检查点抑制剂相关性肺炎(CIP)患者中的应用仍然具有挑战性。虽然越来越多的报道了CIP患者的一线结果,但缺乏后续治疗的有效性和安全性数据。本研究旨在评估一线免疫治疗后既往有CIP的NSCLC患者的治疗模式、生存结果和与二线治疗相关的肺毒性风险。方法:本回顾性研究分析了2019年1月至2024年3月期间在福冈大学医院接受一线联合免疫治疗的159例晚期或复发性NSCLC患者,包括先前接受驱动基因突变分子靶向治疗的患者。根据基线CIP状态对患者进行分层。其中,91例接受二线治疗的患者(19例有CIP病史,72例无CIP病史)进行无进展生存期(PFS)、总生存期(OS)、客观缓解率(ORR)和二线治疗期间药物性肺炎发病情况的评估。结果:32例(20.1%)患者存在基线CIP。既往CIP组转入二线治疗的比例为73%,非既往CIP组为64%,两组间差异无统计学意义(P=0.49),说明既往CIP的存在并没有明显影响接受二线治疗的可能性。二线治疗后的PFS在有和没有先前CIP的患者之间相似。然而,先前CIP组的OS往往更短(6.4 vs 10.3个月),尽管差异无统计学意义(P=0.07),但倾向评分匹配后这种趋势持续存在。在既往有CIP的NSCLC患者中,二线治疗期间药物性肺炎的发生更为频繁(26% vs. 3%)。结论:联合免疫治疗后的二线细胞毒性化疗可能与既往有CIP的NSCLC患者的生存期缩短和肺毒性增加有关。谨慎的临床决策,多学科咨询和密切监测是至关重要的,当开始二线治疗的人群。
{"title":"Analysis of second-line therapy in patients with prior pneumonitis receiving first-line combination immunotherapy for advanced non-small cell lung cancer.","authors":"Noriyuki Ebi, Hiroyuki Inoue, Makoto Fujimoto, Ami Hirata, Maiya Chen, Natsumi Kushima, Yuuka Sakaki, Takato Ikeda, Yuki Shundo, Toyoshi Yanagihara, Masaki Fujita","doi":"10.21037/tlcr-2025-762","DOIUrl":"10.21037/tlcr-2025-762","url":null,"abstract":"<p><strong>Background: </strong>Combination immunotherapy has become a standard first-line treatment for advanced non-small cell lung cancer (NSCLC). However, its application in patients with checkpoint inhibitor-related pneumonitis (CIP) remains challenging due to the risk of treatment-related pulmonary toxicity. While first-line outcomes in CIP patients are increasingly reported, data on the efficacy and safety of subsequent therapies are lacking. This study aimed to evaluate treatment patterns, survival outcomes, and the risk of pulmonary toxicity associated with second-line therapy in NSCLC patients with prior CIP after first-line immunotherapy.</p><p><strong>Methods: </strong>This retrospective study analyzed 159 patients with advanced or recurrent NSCLC treated with first-line combination immunotherapy at Fukuoka University Hospital between January 2019 and March 2024, including those who had previously received molecular targeted therapy for driver gene mutations. Patients were stratified based on baseline CIP status. Among them, 91 patients (19 prior CIP and 72 non-prior CIP) who received second-line therapy were evaluated for progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and drug-induced pneumonitis onset during second-line treatment.</p><p><strong>Results: </strong>Baseline CIP was present in 32 patients (20.1%). The transition rate to second-line therapy was 73% in the prior CIP group and 64% in the non-prior CIP group, with no statistically significant difference observed between the two groups (P=0.49), indicating that the presence of prior CIP did not clearly affect the likelihood of receiving secondary therapy. PFS following second-line treatment was similar between patients with and without prior CIP. However, OS tended to be shorter in the prior CIP group (6.4 <i>vs.</i> 10.3 months), although the difference was not statistically significant (P=0.07), a trend that persisted after propensity score matching. Drug-induced pneumonitis development during second-line therapy occurred more frequently in patients with prior CIP (26% <i>vs.</i> 3%, P<0.01).</p><p><strong>Conclusions: </strong>Second-line cytotoxic chemotherapy following combination immunotherapy may be associated with shorter survival and increased pulmonary toxicity in NSCLC patients with prior CIP. Careful clinical decision-making, multidisciplinary consultation, and close monitoring are essential when initiating second-line treatment in this population.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4210-4220"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605418/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514140","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: Recent studies have increasingly highlighted the critical role of the microbiome in lung cancer initiation, progression, metastasis, and therapeutic resistance. Microbial dysbiosis has been identified as a significant risk factor for lung cancer progression. Beyond the gut, emerging evidence shows that microorganisms also colonize the lungs and even inside tumors, suggesting a broader role of the microbiome in the disease. This review explores the immune-oncology-microbiome (IOM) axis by examining microbial communities across different anatomical sites, their interactions with the tumor immune microenvironment, and their impact on immunotherapy efficacy in lung cancer.
Methods: We conducted a systematic literature search using PubMed and Web of Science databases for English-language articles published between 2014 and 2025, using key terms related to lung cancer and microbiome.
Key content and findings: The microbiome associated with lung cancer encompasses gut microbiota, lower respiratory tract microbiota, and intratumor microbiota. Different microbial communities maintain microecological homeostasis and modulate antitumor immune responses by shaping the tumor immune microenvironment. Meanwhile, certain microbiota can enhance or hinder the effectiveness of immunotherapy.
Conclusions: The IOM axis reveals dynamic interactions between immune cells, cancer cells, and microbes. Specific microbe-derived metabolites show promise as lung cancer biomarkers. Current microbiome-based therapy shows great potential in lung cancer treatment.
背景与目的:近年来的研究越来越强调微生物组在肺癌发生、进展、转移和治疗耐药中的关键作用。微生物生态失调已被确定为肺癌进展的重要危险因素。除了肠道之外,新出现的证据表明,微生物也会在肺部甚至肿瘤内部定居,这表明微生物群在疾病中发挥着更广泛的作用。本文通过研究不同解剖部位的微生物群落、它们与肿瘤免疫微环境的相互作用以及它们对肺癌免疫治疗疗效的影响,探讨了免疫-肿瘤-微生物组(IOM)轴。方法:使用PubMed和Web of Science数据库对2014年至2025年间发表的英文文章进行系统的文献检索,使用与肺癌和微生物组相关的关键术语。关键内容和发现:与肺癌相关的微生物群包括肠道微生物群、下呼吸道微生物群和肿瘤内微生物群。不同的微生物群落通过塑造肿瘤免疫微环境来维持微生态稳态并调节抗肿瘤免疫反应。同时,某些微生物群可以增强或阻碍免疫治疗的有效性。结论:IOM轴揭示了免疫细胞、癌细胞和微生物之间的动态相互作用。特定的微生物衍生代谢物有望成为肺癌的生物标志物。目前基于微生物组的治疗方法在肺癌治疗中显示出巨大的潜力。
{"title":"Immune-oncology-microbiome axis in lung cancer: from carcinogenesis to therapy-a narrative review.","authors":"Mengyi Shen, Yueying Chen, Jiayan Chen, Jie Yin, Chunwei Xu, Dong Wang, Tangfeng Lv","doi":"10.21037/tlcr-2025-737","DOIUrl":"10.21037/tlcr-2025-737","url":null,"abstract":"<p><strong>Background and objective: </strong>Recent studies have increasingly highlighted the critical role of the microbiome in lung cancer initiation, progression, metastasis, and therapeutic resistance. Microbial dysbiosis has been identified as a significant risk factor for lung cancer progression. Beyond the gut, emerging evidence shows that microorganisms also colonize the lungs and even inside tumors, suggesting a broader role of the microbiome in the disease. This review explores the immune-oncology-microbiome (IOM) axis by examining microbial communities across different anatomical sites, their interactions with the tumor immune microenvironment, and their impact on immunotherapy efficacy in lung cancer.</p><p><strong>Methods: </strong>We conducted a systematic literature search using PubMed and Web of Science databases for English-language articles published between 2014 and 2025, using key terms related to lung cancer and microbiome.</p><p><strong>Key content and findings: </strong>The microbiome associated with lung cancer encompasses gut microbiota, lower respiratory tract microbiota, and intratumor microbiota. Different microbial communities maintain microecological homeostasis and modulate antitumor immune responses by shaping the tumor immune microenvironment. Meanwhile, certain microbiota can enhance or hinder the effectiveness of immunotherapy.</p><p><strong>Conclusions: </strong>The IOM axis reveals dynamic interactions between immune cells, cancer cells, and microbes. Specific microbe-derived metabolites show promise as lung cancer biomarkers. Current microbiome-based therapy shows great potential in lung cancer treatment.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4638-4657"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605703/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514233","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 inhibitors (ICIs) are a mainstay in the treatment of non-small cell lung cancer (NSCLC). Although accurate predictive biomarkers are required for treatment selection-considering the risk of serious immune-related adverse events-such predictors remain limited. In this study, we aimed to evaluate the association between the expression of p53, LKB1, and NRF2 proteins and ICI efficacy.
Methods: This retrospective analysis included patients with NSCLC who received first-line ICI-based therapy or epidermal growth factor receptor-tyrosine kinase inhibitors between 2017 and 2025 at our institute. Immunohistochemistry was performed on diagnostic samples to assess p53, LKB1, and NRF2 expression and tumor-infiltrating lymphocytes (TILs). Additionally, the associations between biomarker expression and clinical outcomes were examined.
Results: Among the 43 evaluable cases in the ICI group, aberrant p53 expression was observed in 46.5%, LKB1 loss in 13.9%, and nuclear-dominant NRF2 expression in 34.9% of cases. Aberrant p53 expression was associated with increased TILs and improved progression-free survival (PFS), while the loss of LKB1 was correlated with an immunosuppressive microenvironment and shorter PFS. Nuclear-dominant NRF2 expression was associated with poorer overall survival.
Conclusions: The immunohistochemical profiles of p53 and LKB1 are associated with distinct immune microenvironment features. These markers may serve as surrogate predictors of ICI efficacy in patients with NSCLC.
{"title":"Immunohistochemical analysis of p53 and LKB1 as predictive biomarkers of immune checkpoint inhibitor response in non-small cell lung cancer.","authors":"Kazuo Tsuchiya, Tomo Tsunoda, Hisaki Igarashi, Kazuya Hattori, Taisuke Ito, Takuro Akashi, Yoshiyuki Oyama, Yasuhiko Kitayama, Masaki Ikeda","doi":"10.21037/tlcr-2025-782","DOIUrl":"10.21037/tlcr-2025-782","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) are a mainstay in the treatment of non-small cell lung cancer (NSCLC). Although accurate predictive biomarkers are required for treatment selection-considering the risk of serious immune-related adverse events-such predictors remain limited. In this study, we aimed to evaluate the association between the expression of p53, LKB1, and NRF2 proteins and ICI efficacy.</p><p><strong>Methods: </strong>This retrospective analysis included patients with NSCLC who received first-line ICI-based therapy or epidermal growth factor receptor-tyrosine kinase inhibitors between 2017 and 2025 at our institute. Immunohistochemistry was performed on diagnostic samples to assess p53, LKB1, and NRF2 expression and tumor-infiltrating lymphocytes (TILs). Additionally, the associations between biomarker expression and clinical outcomes were examined.</p><p><strong>Results: </strong>Among the 43 evaluable cases in the ICI group, aberrant p53 expression was observed in 46.5%, LKB1 loss in 13.9%, and nuclear-dominant NRF2 expression in 34.9% of cases. Aberrant p53 expression was associated with increased TILs and improved progression-free survival (PFS), while the loss of LKB1 was correlated with an immunosuppressive microenvironment and shorter PFS. Nuclear-dominant NRF2 expression was associated with poorer overall survival.</p><p><strong>Conclusions: </strong>The immunohistochemical profiles of p53 and LKB1 are associated with distinct immune microenvironment features. These markers may serve as surrogate predictors of ICI efficacy in patients with NSCLC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4527-4540"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12614654/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145542701","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 : 2025-10-31Epub Date: 2025-10-29DOI: 10.21037/tlcr-2025-969
Qihua He, Xin Xu, DongMei Li, Yalei Zhang
Background: RET gene fusions define a distinct molecular subset of non-small cell lung cancer (NSCLC) that commonly affects younger and non-smoking individuals. Selective RET inhibitors such as pralsetinib have shown favorable outcomes in advanced or metastatic settings; however, their role in perioperative or locally advanced disease remains poorly defined. Integrating targeted therapy into a multidisciplinary "sandwich" strategy, which includes neoadjuvant, surgical, and adjuvant components, may offer improved outcomes and requires further exploration.
Case description: We report the case of a 34-year-old male patient with a 15-year smoking history, who presented with persistent right-sided chest pain. Imaging revealed a 3.4 cm × 2.4 cm × 4.4 cm mass in the right middle lobe with mediastinal and left supraclavicular lymphadenopathy. Supraclavicular node biopsy confirmed metastatic poorly differentiated adenocarcinoma. Immunohistochemistry supported a pulmonary origin, and molecular testing via amplification refractory mutation system-polymerase chain reaction (ARMS-PCR) detected a RET exon 12 fusion. The clinical stage was cT2N3M0 (stage IIIB). The patient declined chemotherapy and underwent neoadjuvant pralsetinib therapy. After 3 months, a partial response (PR) was observed. On April 13, 2023, the patient underwent video-assisted thoracoscopic surgery (VATS) with right middle lobectomy. Postoperative pathology revealed a pathological complete response (pCR) with 0% viable tumor cells. Pralsetinib was resumed as adjuvant therapy post-surgery. The patient also began elective regional radiotherapy targeting bilateral supraclavicular drainage areas, but the treatment was terminated early due to grade 3 acute esophagitis. At the 24-month follow-up, the patient remained disease-free on pralsetinib maintenance with preserved performance status.
Conclusions: This case highlights the potential benefit of pralsetinib-based "sandwich" therapy in the treatment of RET fusion-positive NSCLC. Incorporating targeted therapy with surgery and tailored adjuvant treatment may lead to durable remission in selected patients with locally advanced disease. Prospective studies need to be conducted to further define the treatment sequencing and validate this multi-modal approach.
背景:RET基因融合定义了非小细胞肺癌(NSCLC)的一个独特的分子亚群,通常影响年轻人和非吸烟个体。选择性RET抑制剂如普拉塞替尼在晚期或转移性环境中显示出良好的结果;然而,它们在围手术期或局部晚期疾病中的作用仍不明确。将靶向治疗整合到多学科的“三明治”策略中,包括新辅助、手术和辅助成分,可能会提供更好的结果,但需要进一步探索。病例描述:我们报告一例34岁男性患者,吸烟史15年,表现为持续性右侧胸痛。影像学显示右侧中叶3.4 cm × 2.4 cm × 4.4 cm肿块伴纵膈和左侧锁骨上淋巴结病变。锁骨上淋巴结活检证实转移性低分化腺癌。免疫组织化学支持肺起源,通过扩增难治性突变系统-聚合酶链反应(ARMS-PCR)的分子检测检测到RET外显子12融合。临床分期为cT2N3M0 (IIIB期)。患者谢绝化疗,接受新辅助普拉塞替尼治疗。3个月后观察到部分缓解(PR)。2023年4月13日,患者行电视胸腔镜手术(VATS)右中叶切除术。术后病理显示病理完全缓解(pCR),肿瘤细胞存活率为0%。术后恢复普拉塞替尼作为辅助治疗。患者还开始选择性局部放疗,靶向双侧锁骨上引流区,但由于3级急性食管炎,治疗提前终止。在24个月的随访中,患者在普拉塞替尼的维持下保持无病状态,并保持了良好的表现状态。结论:该病例强调了以普拉塞替尼为基础的“三明治”疗法治疗RET融合阳性NSCLC的潜在益处。将靶向治疗与手术和量身定制的辅助治疗相结合,可能导致局部晚期疾病患者的持久缓解。需要进行前瞻性研究以进一步确定治疗顺序并验证这种多模式方法。
{"title":"Pathological complete response in <i>RET</i>-positive non-small cell lung cancer: a case report of pralsetinib-based \"sandwich\" therapy.","authors":"Qihua He, Xin Xu, DongMei Li, Yalei Zhang","doi":"10.21037/tlcr-2025-969","DOIUrl":"10.21037/tlcr-2025-969","url":null,"abstract":"<p><strong>Background: </strong><i>RET</i> gene fusions define a distinct molecular subset of non-small cell lung cancer (NSCLC) that commonly affects younger and non-smoking individuals. Selective <i>RET</i> inhibitors such as pralsetinib have shown favorable outcomes in advanced or metastatic settings; however, their role in perioperative or locally advanced disease remains poorly defined. Integrating targeted therapy into a multidisciplinary \"sandwich\" strategy, which includes neoadjuvant, surgical, and adjuvant components, may offer improved outcomes and requires further exploration.</p><p><strong>Case description: </strong>We report the case of a 34-year-old male patient with a 15-year smoking history, who presented with persistent right-sided chest pain. Imaging revealed a 3.4 cm × 2.4 cm × 4.4 cm mass in the right middle lobe with mediastinal and left supraclavicular lymphadenopathy. Supraclavicular node biopsy confirmed metastatic poorly differentiated adenocarcinoma. Immunohistochemistry supported a pulmonary origin, and molecular testing via amplification refractory mutation system-polymerase chain reaction (ARMS-PCR) detected a <i>RET</i> exon 12 fusion. The clinical stage was cT2N3M0 (stage IIIB). The patient declined chemotherapy and underwent neoadjuvant pralsetinib therapy. After 3 months, a partial response (PR) was observed. On April 13, 2023, the patient underwent video-assisted thoracoscopic surgery (VATS) with right middle lobectomy. Postoperative pathology revealed a pathological complete response (pCR) with 0% viable tumor cells. Pralsetinib was resumed as adjuvant therapy post-surgery. The patient also began elective regional radiotherapy targeting bilateral supraclavicular drainage areas, but the treatment was terminated early due to grade 3 acute esophagitis. At the 24-month follow-up, the patient remained disease-free on pralsetinib maintenance with preserved performance status.</p><p><strong>Conclusions: </strong>This case highlights the potential benefit of pralsetinib-based \"sandwich\" therapy in the treatment of <i>RET</i> fusion-positive NSCLC. Incorporating targeted therapy with surgery and tailored adjuvant treatment may lead to durable remission in selected patients with locally advanced disease. Prospective studies need to be conducted to further define the treatment sequencing and validate this multi-modal approach.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4695-4703"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605663/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145513967","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: Checkpoint inhibitors have revolutionised cancer treatment over the past few decades; however, their successes in clinical trials and real-world settings have been tempered by immune-related adverse events (IrAEs). Checkpoint inhibitor pneumonitis (CIP), the leading cause of IrAE-related mortality, poses a significant challenge to clinicians due to non-specific clinical features and unpredictable disease trajectory. Bronchoalveolar lavage (BAL) is a diagnostic procedure that offers insight into the immune and molecular processes underlying CIP. This review presents the current role and application of BAL in the evaluation of CIP, explores current literature, and discusses how recent advances in research and technology may shape future approaches to its diagnosis and management.
Methods: A comprehensive literature search using PubMed, EMBASE, and Cochrane databases was performed to identify recent literature evaluating BAL findings in CIP. This review synthesizes findings from these studies to provide an up to date and comprehensive overview of the role of BAL in CIP management.
Key content and findings: Current guidelines recommend BAL in symptomatic patients when the diagnosis of CIP remains uncertain, primarily to exclude infection and other disease processes. However, the unpredictable clinical course of CIP narrows the window of opportunity to safely perform BAL. Variable utilisation of BAL in clinical practice may also be attributed to the lack of clinically reliable and applicable biomarkers that could improve diagnostic clarity. Beyond lymphocytosis, a well-recognised finding in BALs of patients with CIP, the role of specific immune cell populations and molecular drivers in shaping a proinflammatory microenvironment has been highlighted in multiple emerging studies. Advances in molecular profiling, immunogenomics, and lung microbiome research hold promise for enhancing our understanding of CIP pathogenesis and guiding future approaches to its diagnosis and management.
Conclusions: In current clinical practice, BAL remains an important investigation to support a diagnosis of CIP; however, its diagnostic value remains uncertain. Enhanced understanding of the CIP immune landscape is fundamental to identifying robust diagnostic and predictive biomarkers that could improve diagnosis and patient outcomes in cancer patients treated with checkpoint inhibitors.
{"title":"Utility of bronchoalveolar lavage in checkpoint inhibitor pneumonitis evaluation: a narrative review.","authors":"Senthuran Shivakumar, Sagun Parakh, Natalia Vukelic, Farzaneh Atashrazm, Vivek Naranbhai, Nicole Soo Leng Goh, Jessica Da Gama Duarte, Tracy Li-Tsein Leong","doi":"10.21037/tlcr-2025-430","DOIUrl":"10.21037/tlcr-2025-430","url":null,"abstract":"<p><strong>Background and objective: </strong>Checkpoint inhibitors have revolutionised cancer treatment over the past few decades; however, their successes in clinical trials and real-world settings have been tempered by immune-related adverse events (IrAEs). Checkpoint inhibitor pneumonitis (CIP), the leading cause of IrAE-related mortality, poses a significant challenge to clinicians due to non-specific clinical features and unpredictable disease trajectory. Bronchoalveolar lavage (BAL) is a diagnostic procedure that offers insight into the immune and molecular processes underlying CIP. This review presents the current role and application of BAL in the evaluation of CIP, explores current literature, and discusses how recent advances in research and technology may shape future approaches to its diagnosis and management.</p><p><strong>Methods: </strong>A comprehensive literature search using PubMed, EMBASE, and Cochrane databases was performed to identify recent literature evaluating BAL findings in CIP. This review synthesizes findings from these studies to provide an up to date and comprehensive overview of the role of BAL in CIP management.</p><p><strong>Key content and findings: </strong>Current guidelines recommend BAL in symptomatic patients when the diagnosis of CIP remains uncertain, primarily to exclude infection and other disease processes. However, the unpredictable clinical course of CIP narrows the window of opportunity to safely perform BAL. Variable utilisation of BAL in clinical practice may also be attributed to the lack of clinically reliable and applicable biomarkers that could improve diagnostic clarity. Beyond lymphocytosis, a well-recognised finding in BALs of patients with CIP, the role of specific immune cell populations and molecular drivers in shaping a proinflammatory microenvironment has been highlighted in multiple emerging studies. Advances in molecular profiling, immunogenomics, and lung microbiome research hold promise for enhancing our understanding of CIP pathogenesis and guiding future approaches to its diagnosis and management.</p><p><strong>Conclusions: </strong>In current clinical practice, BAL remains an important investigation to support a diagnosis of CIP; however, its diagnostic value remains uncertain. Enhanced understanding of the CIP immune landscape is fundamental to identifying robust diagnostic and predictive biomarkers that could improve diagnosis and patient outcomes in cancer patients treated with checkpoint inhibitors.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4628-4637"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605103/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514059","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 : 2025-10-31Epub Date: 2025-10-29DOI: 10.21037/tlcr-2025-599
Apeksha Phulgirkar, Aaleyah Kelso, Rashmikaa Ranganathan, Rebecca N Jerome, Jennifer A Lewis, Lucy B Spalluto
Background: Lung cancer is the leading cause of cancer-related mortality in the United States (U.S.). The United States Preventive Services Task Force (USPSTF) recommends lung cancer screening with low-dose computed tomography (LDCT) for individuals at high risk for lung cancer to improve early detection and survival rates. Despite strong evidence supporting lung cancer screening, its adoption remains low. Participation rates range from only 1% to 16% of those eligible across different geographic regions. Further, rural populations may face significant barriers to lung cancer screening access due to factors such as geographic isolation, socioeconomic status (SES), and health literacy.
Methods: We propose a systematic review to examine how rurality, SES, and health literacy independently and collectively influence lung cancer screening utilization rates in the U.S. Using the Sample, Phenomenon of Interest, Design, Evaluation and Research (SPIDER) framework, this review will synthesize findings from research studies published between the years 2014 to 2024. The review will explore the barriers to screening uptake, including limited healthcare infrastructure, financial constraints, and inadequate awareness or understanding of lung cancer screening guidelines.
Discussion: The findings of this systematic review are intended to influence policies that enhance accessibility, affordability, and education surrounding lung cancer screening to improve outcomes for rural populations.
{"title":"The impact of rurality, socioeconomic status, and health literacy on lung cancer screening uptake: a systematic review protocol.","authors":"Apeksha Phulgirkar, Aaleyah Kelso, Rashmikaa Ranganathan, Rebecca N Jerome, Jennifer A Lewis, Lucy B Spalluto","doi":"10.21037/tlcr-2025-599","DOIUrl":"10.21037/tlcr-2025-599","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is the leading cause of cancer-related mortality in the United States (U.S.). The United States Preventive Services Task Force (USPSTF) recommends lung cancer screening with low-dose computed tomography (LDCT) for individuals at high risk for lung cancer to improve early detection and survival rates. Despite strong evidence supporting lung cancer screening, its adoption remains low. Participation rates range from only 1% to 16% of those eligible across different geographic regions. Further, rural populations may face significant barriers to lung cancer screening access due to factors such as geographic isolation, socioeconomic status (SES), and health literacy.</p><p><strong>Methods: </strong>We propose a systematic review to examine how rurality, SES, and health literacy independently and collectively influence lung cancer screening utilization rates in the U.S. Using the Sample, Phenomenon of Interest, Design, Evaluation and Research (SPIDER) framework, this review will synthesize findings from research studies published between the years 2014 to 2024. The review will explore the barriers to screening uptake, including limited healthcare infrastructure, financial constraints, and inadequate awareness or understanding of lung cancer screening guidelines.</p><p><strong>Discussion: </strong>The findings of this systematic review are intended to influence policies that enhance accessibility, affordability, and education surrounding lung cancer screening to improve outcomes for rural populations.</p><p><strong>Registration: </strong>PROSPERO Registration: CRD42025644774.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4658-4665"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605193/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514078","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: Current standard of care for squamous non-small cell lung cancer (sqNSCLC) is not very effective, and FGFR2b-targeted therapy may be a new option for patients with sqNSCLC. The prevalence and clinical implications of fibroblast growth factor receptor 2b (FGFR2b) expression in Chinese patients with advanced sqNSCLC remain poorly characterized. This study evaluated FGFR2b expression patterns, their association with programmed death-ligand 1 (PD-L1) expression, and clinical outcomes in this population.
Methods: This retrospective cross-sectional analysis screened three hundred newly diagnosed advanced sqNSCLC patients from 2018 to 2024. Archival biopsy samples were assessed for FGFR2b expression via immunohistochemistry (IHC). A total of 266 patients, with histologically confirmed stage IIIB-IV sqNSCLC and valid FGFR2b protein expression status, were enrolled. Historical clinical data and PD-L1 detection reports were extracted from hospital records.
Results: Among the 266 patients, 117 (44.0%, 117/266) were diagnosed with IIIB/IIIC-stage cancer, and 149 (56%, 149/266) were diagnosed with IV-stage cancer. FGFR2b positivity (≥1+ in ≥1% tumor cells) was observed in 50.8% of patients, while overexpression (≥2+ in ≥1% tumor cells) occurred in 19.5%. FGFR2b expression was correlated significantly with sex (P=0.04), and cancer stage (P=0.01). Among 204 patients with valid FGFR2b and PD-L1 dual expression data, over 80% of FGFR2b-positive tumors demonstrated concurrent PD-L1 expression. In patients receiving first-line paclitaxel/nab-paclitaxel-platinum combined with anti-programmed cell death-1 (PD-1)/PD-L1 antibodies, FGFR2b-positive cohorts exhibited superior outcomes: higher objective response rate (57.4% vs. 41.9%) and prolonged median progression-free survival {11.3 months [95% confidence interval (CI): 6.6-17.9] vs. 6.5 months (95% CI: 5.0-17.3)} compared to FGFR2b-negative counterparts.
Conclusions: FGFR2b expression may serve as a predictive biomarker for therapeutic response of FGFR2b-targeted agents in sqNSCLC. The high co-expression rate of FGFR2b and PD-L1 supports the feasibility of combining FGFR2b-targeted agents with immune checkpoint inhibitors as a rational treatment strategy.
{"title":"A retrospective cross-sectional study of FGFR2b alterations in Chinese patients with advanced squamous non-small cell lung cancer: prevalence, programmed death-ligand 1 co-expression patterns, and association with clinical outcomes.","authors":"Xiyuan Wang, Kaiwen Chi, Haiyue Wang, Shuguang Sun, Chunxia Ao, Jiongyan Li, Yuanjie Ge, Chen Yang, Yayuan Fu, Dongmei Lin, Renhong Tang, Wei Sun","doi":"10.21037/tlcr-2025-452","DOIUrl":"10.21037/tlcr-2025-452","url":null,"abstract":"<p><strong>Background: </strong>Current standard of care for squamous non-small cell lung cancer (sqNSCLC) is not very effective, and FGFR2b-targeted therapy may be a new option for patients with sqNSCLC. The prevalence and clinical implications of fibroblast growth factor receptor 2b (FGFR2b) expression in Chinese patients with advanced sqNSCLC remain poorly characterized. This study evaluated FGFR2b expression patterns, their association with programmed death-ligand 1 (PD-L1) expression, and clinical outcomes in this population.</p><p><strong>Methods: </strong>This retrospective cross-sectional analysis screened three hundred newly diagnosed advanced sqNSCLC patients from 2018 to 2024. Archival biopsy samples were assessed for FGFR2b expression via immunohistochemistry (IHC). A total of 266 patients, with histologically confirmed stage IIIB-IV sqNSCLC and valid FGFR2b protein expression status, were enrolled. Historical clinical data and PD-L1 detection reports were extracted from hospital records.</p><p><strong>Results: </strong>Among the 266 patients, 117 (44.0%, 117/266) were diagnosed with IIIB/IIIC-stage cancer, and 149 (56%, 149/266) were diagnosed with IV-stage cancer. FGFR2b positivity (≥1+ in ≥1% tumor cells) was observed in 50.8% of patients, while overexpression (≥2+ in ≥1% tumor cells) occurred in 19.5%. FGFR2b expression was correlated significantly with sex (P=0.04), and cancer stage (P=0.01). Among 204 patients with valid FGFR2b and PD-L1 dual expression data, over 80% of FGFR2b-positive tumors demonstrated concurrent PD-L1 expression. In patients receiving first-line paclitaxel/nab-paclitaxel-platinum combined with anti-programmed cell death-1 (PD-1)/PD-L1 antibodies, FGFR2b-positive cohorts exhibited superior outcomes: higher objective response rate (57.4% <i>vs.</i> 41.9%) and prolonged median progression-free survival {11.3 months [95% confidence interval (CI): 6.6-17.9] <i>vs.</i> 6.5 months (95% CI: 5.0-17.3)} compared to FGFR2b-negative counterparts.</p><p><strong>Conclusions: </strong>FGFR2b expression may serve as a predictive biomarker for therapeutic response of FGFR2b-targeted agents in sqNSCLC. The high co-expression rate of FGFR2b and PD-L1 supports the feasibility of combining FGFR2b-targeted agents with immune checkpoint inhibitors as a rational treatment strategy.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 10","pages":"4371-4383"},"PeriodicalIF":3.5,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12605099/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145514120","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}