Pub Date : 2025-11-30Epub Date: 2025-11-27DOI: 10.21037/tlcr-2025-940
Jihwan Choi, Jooyong Shim, Jonghoon Kim, Yeon Jeong Kim, Changha Hwang, Woong-Yang Park, Jae Myoung Noh, Hongryull Pyo, Ho Yun Lee
Background: Liquid biopsy based on cell-free DNA (cfDNA) in oncology has emerged as a promising technique for tracking cancer dynamics, especially for detecting minimal residual disease. To date, most studies have used cfDNA for static evaluations of tumor burden. In this study, we propose a novel approach integrating serial cfDNA and computed tomography (CT) tumor volume to fully reflect the dynamic nature of tumor response after treatment.
Methods: This prospective study involved 25 patients treated with curative-intent radiotherapy for localized non-small cell lung cancer (NSCLC) between June 2019 and November 2020, with 17 subsequently included in final analysis. Longitudinal blood samples were divided into two phases relative to day 3 after treatment initiation, and kinetic parameters, such as velocity and acceleration of cfDNA levels, were calculated. To complement sparse samplings in later days, volume data from routine CT scans were incorporated. K-means clustering using two different variable sets (cfDNA only and cfDNA with volume parameters) and conventional assessment using Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 were applied to stratify patients, and their performance was compared.
Results: The model incorporating both cfDNA and volume parameters effectively separated responders (mean progression-free survival, 44.2 months) from non-responders [16.6 months, P=0.02; area under the receiver operating characteristic curve (AUC) =0.955], outperforming cfDNA only model (36.0 vs. 14.5 months, P=0.04; AUC =0.848). In contrast, RECIST v1.1-based conventional assessment showed no significant difference (P=0.62, AUC =0.70).
Conclusions: Therefore, our study demonstrates that integration of longitudinal cfDNA and tumor volume dynamics yielded improved assessment of treatment response and prognosis in NSCLC.
{"title":"Integrative modeling of longitudinal cell-free DNA and tumor volume dynamics: a multimodal quantitative prognostic framework.","authors":"Jihwan Choi, Jooyong Shim, Jonghoon Kim, Yeon Jeong Kim, Changha Hwang, Woong-Yang Park, Jae Myoung Noh, Hongryull Pyo, Ho Yun Lee","doi":"10.21037/tlcr-2025-940","DOIUrl":"10.21037/tlcr-2025-940","url":null,"abstract":"<p><strong>Background: </strong>Liquid biopsy based on cell-free DNA (cfDNA) in oncology has emerged as a promising technique for tracking cancer dynamics, especially for detecting minimal residual disease. To date, most studies have used cfDNA for static evaluations of tumor burden. In this study, we propose a novel approach integrating serial cfDNA and computed tomography (CT) tumor volume to fully reflect the dynamic nature of tumor response after treatment.</p><p><strong>Methods: </strong>This prospective study involved 25 patients treated with curative-intent radiotherapy for localized non-small cell lung cancer (NSCLC) between June 2019 and November 2020, with 17 subsequently included in final analysis. Longitudinal blood samples were divided into two phases relative to day 3 after treatment initiation, and kinetic parameters, such as velocity and acceleration of cfDNA levels, were calculated. To complement sparse samplings in later days, volume data from routine CT scans were incorporated. K-means clustering using two different variable sets (cfDNA only and cfDNA with volume parameters) and conventional assessment using Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 were applied to stratify patients, and their performance was compared.</p><p><strong>Results: </strong>The model incorporating both cfDNA and volume parameters effectively separated responders (mean progression-free survival, 44.2 months) from non-responders [16.6 months, P=0.02; area under the receiver operating characteristic curve (AUC) =0.955], outperforming cfDNA only model (36.0 <i>vs.</i> 14.5 months, P=0.04; AUC =0.848). In contrast, RECIST v1.1-based conventional assessment showed no significant difference (P=0.62, AUC =0.70).</p><p><strong>Conclusions: </strong>Therefore, our study demonstrates that integration of longitudinal cfDNA and tumor volume dynamics yielded improved assessment of treatment response and prognosis in NSCLC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4746-4755"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715792","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) have transformed the treatment landscape for advanced non-small cell lung cancer (NSCLC). However, their efficacy and safety in patients with idiopathic interstitial pneumonias (IIPs) or interstitial lung abnormalities (ILAs) remain poorly defined, as such patients have been largely excluded from pivotal clinical trials. This study aimed to assess real-world outcomes and adverse events associated with ICI-based therapies in patients with NSCLC and coexisting IIPs or ILAs.
Methods: We conducted a multicenter retrospective cohort study across 11 institutions affiliated with the North Japan Lung Cancer Study Group. Patients with advanced NSCLC and pre-existing IIPs or ILAs who received ICI monotherapy or ICI plus chemotherapy (chemo-ICI) between December 2015 and March 2020 were included. Data on progression-free survival (PFS), overall survival (OS), treatment response, and pneumonitis incidence were collected. High-resolution computed tomography (HRCT) images were centrally reviewed to classify the radiologic patterns of IIPs and ILAs.
Results: A total of 53 patients with IIPs and 18 with ILAs were analyzed. In the IIP cohort, the median PFS was 5.1 months with chemo-ICI, 7.3 months with first-line ICI monotherapy, and 5.7 months with second-line or later ICI monotherapy. The median OS ranged from 5.4 to 15.4 months, depending on the treatment type. Grade 3-4 pneumonitis occurred in 17.0% of patients with IIPs. In the ILA cohort, chemo-ICI resulted in a median OS of 27.0 months with no cases of pneumonitis, while ICI monotherapy was associated with a 22.2% pneumonitis incidence. Subpleural fibrotic ILAs were linked to a higher risk of pneumonitis. Long-term survivors were identified in both cohorts.
Conclusions: ICI therapy may provide durable survival benefits for patients with NSCLC and underlying IIPs or ILAs. However, the elevated incidence of high-grade pneumonitis-particularly in patients with IIPs or fibrotic ILAs-underscores the importance of careful patient selection. Prospective studies are needed to refine treatment strategies in this population.
{"title":"Efficacy and safety of immune checkpoint inhibitors in advanced non-small cell lung cancer with idiopathic interstitial pneumonia or interstitial lung abnormalities: NJLCG2301.","authors":"Fumihiko Okumura, Hisashi Tanaka, Katsuhiro Onodera, Ryo Morita, Minehiko Inomata, Aya Suzuki, Jun Sugisaka, Hiroyuki Minemura, Daisuke Jingu, Satoshi Kudo, Hajime Kikuchi, Naruo Yoshimura, Fumiyasu Tsushima, Shingo Kakeda, Sadatomo Tasaka","doi":"10.21037/tlcr-2025-783","DOIUrl":"10.21037/tlcr-2025-783","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape for advanced non-small cell lung cancer (NSCLC). However, their efficacy and safety in patients with idiopathic interstitial pneumonias (IIPs) or interstitial lung abnormalities (ILAs) remain poorly defined, as such patients have been largely excluded from pivotal clinical trials. This study aimed to assess real-world outcomes and adverse events associated with ICI-based therapies in patients with NSCLC and coexisting IIPs or ILAs.</p><p><strong>Methods: </strong>We conducted a multicenter retrospective cohort study across 11 institutions affiliated with the North Japan Lung Cancer Study Group. Patients with advanced NSCLC and pre-existing IIPs or ILAs who received ICI monotherapy or ICI plus chemotherapy (chemo-ICI) between December 2015 and March 2020 were included. Data on progression-free survival (PFS), overall survival (OS), treatment response, and pneumonitis incidence were collected. High-resolution computed tomography (HRCT) images were centrally reviewed to classify the radiologic patterns of IIPs and ILAs.</p><p><strong>Results: </strong>A total of 53 patients with IIPs and 18 with ILAs were analyzed. In the IIP cohort, the median PFS was 5.1 months with chemo-ICI, 7.3 months with first-line ICI monotherapy, and 5.7 months with second-line or later ICI monotherapy. The median OS ranged from 5.4 to 15.4 months, depending on the treatment type. Grade 3-4 pneumonitis occurred in 17.0% of patients with IIPs. In the ILA cohort, chemo-ICI resulted in a median OS of 27.0 months with no cases of pneumonitis, while ICI monotherapy was associated with a 22.2% pneumonitis incidence. Subpleural fibrotic ILAs were linked to a higher risk of pneumonitis. Long-term survivors were identified in both cohorts.</p><p><strong>Conclusions: </strong>ICI therapy may provide durable survival benefits for patients with NSCLC and underlying IIPs or ILAs. However, the elevated incidence of high-grade pneumonitis-particularly in patients with IIPs or fibrotic ILAs-underscores the importance of careful patient selection. Prospective studies are needed to refine treatment strategies in this population.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4756-4767"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683449/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715804","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-2025-894
Tong Li, Yang Zhang, Hong Hu, Ting Ye, Yihua Sun, Yawei Zhang, Jiaqing Xiang, Haiquan Chen
Background: While most patients with stage I non-small cell lung cancer (NSCLC) remain recurrence-free after resection, some still develop recurrent disease. The surgical curative time window concept, defined as no recurrence through 5-year follow-up, helps identify potentially cured patients, yet predictive clinicopathologic features in stage I invasive NSCLC need clarification. This study sought to identify such features to enable risk-adapted surveillance.
Methods: We analyzed a prospectively collected dataset of patients with stage I invasive NSCLC who underwent R0 resection between 2008 and 2015. Cox regression analysis was used to evaluate the association between clinicopathologic features and disease recurrence, aiming to identify independent prognostic factors.
Results: A total of 1,817 patients met the inclusion criteria. The 5-year cumulative incidence of recurrence was 14.6%. Female sex, tumor size ≤2 cm, lepidic-predominant adenocarcinoma (LPA) histologic type, presence of a ground-glass opacity (GGO) component, and solid component size ≤10 mm were identified as independent prognostic factors. A risk stratification system was subsequently developed, classifying patients into two groups: a low-risk group (with ≥4 factors; n=341) and an elevated-risk group (with <4 factors; n=1,476). Kaplan-Meier analysis revealed statistically significant differences in recurrence-free survival (RFS), overall survival (OS), and lung cancer-specific survival (LCSS) between the two groups (P<0.001). The low-risk group is considered to represent the population within the surgical curative time window.
Conclusions: Patients with stage I invasive NSCLC who meet at least four of the following five criteria-female sex, tumor size ≤2 cm, solid component ≤10 mm, presence of a GGO component, and LPA histologic type-may be considered within the "surgical curative time window" and may therefore qualify for reduced surveillance intensity.
{"title":"Defining the surgical curative time window: identifying patients with an absence of recurrence for 5 years following surgical resection of stage I invasive non-small cell lung cancer.","authors":"Tong Li, Yang Zhang, Hong Hu, Ting Ye, Yihua Sun, Yawei Zhang, Jiaqing Xiang, Haiquan Chen","doi":"10.21037/tlcr-2025-894","DOIUrl":"10.21037/tlcr-2025-894","url":null,"abstract":"<p><strong>Background: </strong>While most patients with stage I non-small cell lung cancer (NSCLC) remain recurrence-free after resection, some still develop recurrent disease. The surgical curative time window concept, defined as no recurrence through 5-year follow-up, helps identify potentially cured patients, yet predictive clinicopathologic features in stage I invasive NSCLC need clarification. This study sought to identify such features to enable risk-adapted surveillance.</p><p><strong>Methods: </strong>We analyzed a prospectively collected dataset of patients with stage I invasive NSCLC who underwent R0 resection between 2008 and 2015. Cox regression analysis was used to evaluate the association between clinicopathologic features and disease recurrence, aiming to identify independent prognostic factors.</p><p><strong>Results: </strong>A total of 1,817 patients met the inclusion criteria. The 5-year cumulative incidence of recurrence was 14.6%. Female sex, tumor size ≤2 cm, lepidic-predominant adenocarcinoma (LPA) histologic type, presence of a ground-glass opacity (GGO) component, and solid component size ≤10 mm were identified as independent prognostic factors. A risk stratification system was subsequently developed, classifying patients into two groups: a low-risk group (with ≥4 factors; n=341) and an elevated-risk group (with <4 factors; n=1,476). Kaplan-Meier analysis revealed statistically significant differences in recurrence-free survival (RFS), overall survival (OS), and lung cancer-specific survival (LCSS) between the two groups (P<0.001). The low-risk group is considered to represent the population within the surgical curative time window.</p><p><strong>Conclusions: </strong>Patients with stage I invasive NSCLC who meet at least four of the following five criteria-female sex, tumor size ≤2 cm, solid component ≤10 mm, presence of a GGO component, and LPA histologic type-may be considered within the \"surgical curative time window\" and may therefore qualify for reduced surveillance intensity.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4719-4732"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683451/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715636","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-2025-414
Mary E Gwin, Mitchell S von Itzstein, Jialiang Liu, Farjana J Fattah, Hong Mu-Mosley, Jeffrey A SoRelle, Sheena Bhalla, Jonathan E Dowell, Sawsan Rashdan, Jason Y Park, Donghan M Yang, Yang Xie, David E Gerber
Background: In non-small cell lung cancer (NSCLC), programmed death-ligand 1 (PD-L1) expression has moderate ability to predict immune checkpoint inhibitor (ICI) benefit. In clinical practice, PD-L1, a cell surface protein, cannot be characterized in currently available blood-based tests such as circulating tumor DNA assays. To understand the biologic effects of PD-L1 more fully and evaluate whether blood-based tests could provide insight into its expression, we determined the association between PD-L1 expression and systemic immune parameters.
Methods: We collected pre- and post-treatment (6-week) peripheral blood samples in patients with NSCLC treated with ICI. Using multiplex panels and cytometry by time of flight (CyTOF), we analyzed specimens for baseline and post-treatment changes in cytokines, autoantibodies, and immune cell populations. We determined the association between case characteristics, immune parameters, and tumor PD-L1 expression (categorized as <1%, 1-49%, and ≥50%) using Chi-square, one-way analysis of variance (ANOVA), and Kruskal-Wallis tests, accounting for multiple comparisons.
Results: A total of 119 patients were included in the analysis, of whom 41 (34%) had PD-L1 expression <1%; 44 (37%), 1-49%; and 34 (29%), ≥50%. PD-L1 expression was not associated with any demographic, tumor, or treatment characteristics. Among 39 cytokines evaluated, baseline levels of macrophage migration inhibitory factor (MIF) were significantly greater in high PD-L1 positive cases. Among 124 autoantibodies included in the analysis, three (anti-aggrecan, -proteoglycan, and -nucleosome) demonstrated significantly greater post-ICI treatment increases in cases with higher PD-L1 expression. In PD-L1 positive cases, baseline abundance of natural killer T (NKT) cells (P=0.001) and activated monocytes (P=0.04) were significantly lower, while post-treatment increases in mature natural killer (NK) cells were significantly greater (P=0.006).
Conclusions: NSCLC PD-L1 expression is associated with few systemic immune parameters, suggesting that effects on anti-tumor immunity may occur predominantly in the tumor microenvironment and that blood-based assays are unlikely to provide meaningful surrogates of this biomarker.
{"title":"Association of PD-L1 expression with systemic immune parameters in non-small cell lung cancer.","authors":"Mary E Gwin, Mitchell S von Itzstein, Jialiang Liu, Farjana J Fattah, Hong Mu-Mosley, Jeffrey A SoRelle, Sheena Bhalla, Jonathan E Dowell, Sawsan Rashdan, Jason Y Park, Donghan M Yang, Yang Xie, David E Gerber","doi":"10.21037/tlcr-2025-414","DOIUrl":"10.21037/tlcr-2025-414","url":null,"abstract":"<p><strong>Background: </strong>In non-small cell lung cancer (NSCLC), programmed death-ligand 1 (PD-L1) expression has moderate ability to predict immune checkpoint inhibitor (ICI) benefit. In clinical practice, PD-L1, a cell surface protein, cannot be characterized in currently available blood-based tests such as circulating tumor DNA assays. To understand the biologic effects of PD-L1 more fully and evaluate whether blood-based tests could provide insight into its expression, we determined the association between PD-L1 expression and systemic immune parameters.</p><p><strong>Methods: </strong>We collected pre- and post-treatment (6-week) peripheral blood samples in patients with NSCLC treated with ICI. Using multiplex panels and cytometry by time of flight (CyTOF), we analyzed specimens for baseline and post-treatment changes in cytokines, autoantibodies, and immune cell populations. We determined the association between case characteristics, immune parameters, and tumor PD-L1 expression (categorized as <1%, 1-49%, and ≥50%) using Chi-square, one-way analysis of variance (ANOVA), and Kruskal-Wallis tests, accounting for multiple comparisons.</p><p><strong>Results: </strong>A total of 119 patients were included in the analysis, of whom 41 (34%) had PD-L1 expression <1%; 44 (37%), 1-49%; and 34 (29%), ≥50%. PD-L1 expression was not associated with any demographic, tumor, or treatment characteristics. Among 39 cytokines evaluated, baseline levels of macrophage migration inhibitory factor (MIF) were significantly greater in high PD-L1 positive cases. Among 124 autoantibodies included in the analysis, three (anti-aggrecan, -proteoglycan, and -nucleosome) demonstrated significantly greater post-ICI treatment increases in cases with higher PD-L1 expression. In PD-L1 positive cases, baseline abundance of natural killer T (NKT) cells (P=0.001) and activated monocytes (P=0.04) were significantly lower, while post-treatment increases in mature natural killer (NK) cells were significantly greater (P=0.006).</p><p><strong>Conclusions: </strong>NSCLC PD-L1 expression is associated with few systemic immune parameters, suggesting that effects on anti-tumor immunity may occur predominantly in the tumor microenvironment and that blood-based assays are unlikely to provide meaningful surrogates of this biomarker.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4962-4972"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715878","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: Lung cancer is a leading cause of cancer-related mortality worldwide, with heterogeneity and acquired resistance posing major challenges to treatment. Advances in next-generation sequencing (NGS) have enabled comprehensive genomic profiling, yet there remains a need for robust patient-derived models to study tumor biology and inform precision medicine. This study aims to establish and characterize patient-derived lung cancer organoids (LCOs) using whole-genome sequencing (WGS) to explore their genomic landscape and therapeutic potential.
Methods: We established a panel of LCOs from resected tumors and malignant pleural effusions (MPEs) of 14 non-small cell lung cancer (NSCLC) patients. Organoids were authenticated and subjected to WGS to profile somatic single nucleotide variants (SNVs), insertions/deletions (InDels), copy number variations (CNVs), structural variants (SVs), and microsatellite instability (MSI). Bioinformatic analyses were performed to annotate mutations, assess tumor mutation burden (TMB), and explore mutational signatures. Furthermore, deep learning-based drug response prediction and in vitro drug sensitivity assays were conducted to evaluate therapeutic potentials in the established LCOs.
Results: In the established LCOs, WGS revealed recurrent mutations in TP53, TTN, MUC16, and FLG, with approximately 80% of somatic variants located in non-coding regions, highlighting the potential role of regulatory elements in lung cancer pathogenesis. Early and locally advanced-stage tumor-derived LCOs exhibited higher TMB and MSI compared to those from advanced-stage disease, suggesting greater clonal diversity prior to therapeutic intervention. Drug screening demonstrated the feasibility of using genomic data for drug prediction, but requires more advanced models to fully utilize the WGS data.
Conclusions: Our comprehensive genomic characterization of patient-derived LCOs provides valuable insights into the mutational landscape and evolutionary dynamics of lung cancer. These well-annotated organoid models serve as a powerful resource for investigating tumor biology and developing genomically informed therapeutic strategies.
{"title":"Whole genome characterization of patient-derived lung cancer organoids.","authors":"Hoi-Hin Kwok, Nerissa Chui-Mei Lee, Junyang Deng, Jiashuang Yang, Lynn Yim-Wah Shong, Cally Ka-Lai Ho, Kwok-Fai Lee, Michael Kuan-Yew Hsin, Hongjing Zang, Joshua Jing-Xi Li, David Chi-Leung Lam","doi":"10.21037/tlcr-2025-738","DOIUrl":"10.21037/tlcr-2025-738","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is a leading cause of cancer-related mortality worldwide, with heterogeneity and acquired resistance posing major challenges to treatment. Advances in next-generation sequencing (NGS) have enabled comprehensive genomic profiling, yet there remains a need for robust patient-derived models to study tumor biology and inform precision medicine. This study aims to establish and characterize patient-derived lung cancer organoids (LCOs) using whole-genome sequencing (WGS) to explore their genomic landscape and therapeutic potential.</p><p><strong>Methods: </strong>We established a panel of LCOs from resected tumors and malignant pleural effusions (MPEs) of 14 non-small cell lung cancer (NSCLC) patients. Organoids were authenticated and subjected to WGS to profile somatic single nucleotide variants (SNVs), insertions/deletions (InDels), copy number variations (CNVs), structural variants (SVs), and microsatellite instability (MSI). Bioinformatic analyses were performed to annotate mutations, assess tumor mutation burden (TMB), and explore mutational signatures. Furthermore, deep learning-based drug response prediction and in vitro drug sensitivity assays were conducted to evaluate therapeutic potentials in the established LCOs.</p><p><strong>Results: </strong>In the established LCOs, WGS revealed recurrent mutations in <i>TP53</i>, <i>TTN</i>, <i>MUC16</i>, and <i>FLG</i>, with approximately 80% of somatic variants located in non-coding regions, highlighting the potential role of regulatory elements in lung cancer pathogenesis. Early and locally advanced-stage tumor-derived LCOs exhibited higher TMB and MSI compared to those from advanced-stage disease, suggesting greater clonal diversity prior to therapeutic intervention. Drug screening demonstrated the feasibility of using genomic data for drug prediction, but requires more advanced models to fully utilize the WGS data.</p><p><strong>Conclusions: </strong>Our comprehensive genomic characterization of patient-derived LCOs provides valuable insights into the mutational landscape and evolutionary dynamics of lung cancer. These well-annotated organoid models serve as a powerful resource for investigating tumor biology and developing genomically informed therapeutic strategies.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4906-4922"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683417/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715948","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-2025-1133
Haoran Zhang, Zhaozhen Wu, Tao Lu, Hui Zhang, Ji Li, Dongming Zhang, Siyuan Yu, Xiaobei Guo, Xiaoyi Feng, Jia Liu, Yuequan Shi, Xiaoxing Gao, Xiaoyan Liu, Minjiang Chen, Jing Zhao, Wei Zhong, Ying Hu, Yan Xu, Mengzhao Wang
Background: Clinical trials have demonstrated the efficacy of both alectinib and crizotinib in anaplastic lymphoma kinase (ALK) fusion (ALK-positive) non-small cell lung cancer (NSCLC). However, a critical question persists regarding their real-world comparative effectiveness in Chinese patients, especially in light of divergent overall survival (OS) results from Western and Asian trials. To address this uncertainty, this real-world study aimed to directly compare the efficacy of alectinib and crizotinib as first-line therapies in a Chinese clinical setting.
Methods: Patients diagnosed with ALK-positive NSCLC from two centers in China were included in the study. Progression-free survival (PFS) was the primary endpoint, while OS, the disease control rate, the objective response rate, and safety were the secondary endpoints. Second-line therapy and prognostic factors were also investigated.
Results: In total, 261 treatment-naïve patients were evaluated, of whom, 128 received crizotinib, and 133 received alectinib. Compared with crizotinib, alectinib demonstrated significant superiority in terms of PFS [45.5 vs. 16.6 months, hazard ratio (HR) =0.36, P<0.001]. However, the PFS of the patients who received first-line alectinib therapy was comparable to that of the patients who received both first-line crizotinib therapy and second-line therapy (45.5 vs. 43.9 months, P=0.32). Similarly, the OS was comparable between the alectinib and crizotinib treatment groups (alectinib vs. crizotinib, not reached vs. 71.2 months, P=0.23). The alectinib treatment group also had a lower incidence of adverse events (AEs) than the crizotinib treatment group (34.6% vs. 50.0%, P=0.01). The occurrence rates of grade 3 or higher AEs were comparable between the two groups (alectinib vs. crizotinib, 4.5% vs. 5.5%). Further, receiving crizotinib instead of alectinib as the first-line therapy and the presence of bone and adrenal metastases at the baseline were independent risk factors for PFS. While the presence of bone, liver, and adrenal metastasis were independent risk factors for OS.
Conclusions: Alectinib is recommended over crizotinib in the treatment of patients with ALK-positive NSCLC.
背景:临床试验已经证明了阿勒替尼和克唑替尼治疗间变性淋巴瘤激酶(ALK)融合(ALK阳性)非小细胞肺癌(NSCLC)的疗效。然而,一个关键的问题仍然存在,即它们在中国患者中的实际比较有效性,特别是考虑到西方和亚洲试验的总生存期(OS)结果不同。为了解决这一不确定性,这项现实世界的研究旨在直接比较阿勒替尼和克唑替尼作为一线治疗在中国临床环境中的疗效。方法:将中国两个中心诊断为alk阳性NSCLC的患者纳入研究。无进展生存期(PFS)是主要终点,而OS、疾病控制率、客观缓解率和安全性是次要终点。二线治疗和预后因素也进行了调查。结果:共评估261例treatment-naïve患者,其中128例接受克唑替尼治疗,133例接受阿勒替尼治疗。与克唑替尼相比,阿勒替尼在PFS方面表现出显著优势[45.5 vs. 16.6个月,风险比(HR) =0.36, PFS = 43.9个月,P=0.32]。同样,阿勒替尼治疗组和克唑替尼治疗组之间的OS具有可比性(阿勒替尼vs克唑替尼,未达到vs 71.2个月,P=0.23)。阿勒替尼治疗组不良事件发生率(ae)也低于克唑替尼治疗组(34.6%比50.0%,P=0.01)。两组间3级及以上ae的发生率具有可比性(阿勒替尼vs克唑替尼,4.5% vs 5.5%)。此外,接受克唑替尼而不是阿勒替尼作为一线治疗以及基线时骨和肾上腺转移的存在是PFS的独立危险因素。而骨、肝和肾上腺转移的存在是OS的独立危险因素。结论:alk阳性NSCLC患者推荐使用Alectinib而不是crizotinib。
{"title":"Alectinib versus crizotinib as the first-line treatment in patients with advanced ALK-positive non-small cell lung cancer: a Chinese real-world cohort study.","authors":"Haoran Zhang, Zhaozhen Wu, Tao Lu, Hui Zhang, Ji Li, Dongming Zhang, Siyuan Yu, Xiaobei Guo, Xiaoyi Feng, Jia Liu, Yuequan Shi, Xiaoxing Gao, Xiaoyan Liu, Minjiang Chen, Jing Zhao, Wei Zhong, Ying Hu, Yan Xu, Mengzhao Wang","doi":"10.21037/tlcr-2025-1133","DOIUrl":"10.21037/tlcr-2025-1133","url":null,"abstract":"<p><strong>Background: </strong>Clinical trials have demonstrated the efficacy of both alectinib and crizotinib in anaplastic lymphoma kinase (ALK) fusion (ALK-positive) non-small cell lung cancer (NSCLC). However, a critical question persists regarding their real-world comparative effectiveness in Chinese patients, especially in light of divergent overall survival (OS) results from Western and Asian trials. To address this uncertainty, this real-world study aimed to directly compare the efficacy of alectinib and crizotinib as first-line therapies in a Chinese clinical setting.</p><p><strong>Methods: </strong>Patients diagnosed with ALK-positive NSCLC from two centers in China were included in the study. Progression-free survival (PFS) was the primary endpoint, while OS, the disease control rate, the objective response rate, and safety were the secondary endpoints. Second-line therapy and prognostic factors were also investigated.</p><p><strong>Results: </strong>In total, 261 treatment-naïve patients were evaluated, of whom, 128 received crizotinib, and 133 received alectinib. Compared with crizotinib, alectinib demonstrated significant superiority in terms of PFS [45.5 <i>vs.</i> 16.6 months, hazard ratio (HR) =0.36, P<0.001]. However, the PFS of the patients who received first-line alectinib therapy was comparable to that of the patients who received both first-line crizotinib therapy and second-line therapy (45.5 <i>vs.</i> 43.9 months, P=0.32). Similarly, the OS was comparable between the alectinib and crizotinib treatment groups (alectinib <i>vs.</i> crizotinib, not reached <i>vs.</i> 71.2 months, P=0.23). The alectinib treatment group also had a lower incidence of adverse events (AEs) than the crizotinib treatment group (34.6% <i>vs.</i> 50.0%, P=0.01). The occurrence rates of grade 3 or higher AEs were comparable between the two groups (alectinib <i>vs.</i> crizotinib, 4.5% <i>vs.</i> 5.5%). Further, receiving crizotinib instead of alectinib as the first-line therapy and the presence of bone and adrenal metastases at the baseline were independent risk factors for PFS. While the presence of bone, liver, and adrenal metastasis were independent risk factors for OS.</p><p><strong>Conclusions: </strong>Alectinib is recommended over crizotinib in the treatment of patients with ALK-positive NSCLC.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5044-5058"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683480/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715790","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: 2025-11-26DOI: 10.21037/tlcr-2025-1075
Qiang Wu, Li Wang, Hao Su, Ting Lei
{"title":"A new hope for aggressive thoracic cancers: foundational efficacy of frontline immunotherapy in SMARCA4-deficient tumors.","authors":"Qiang Wu, Li Wang, Hao Su, Ting Lei","doi":"10.21037/tlcr-2025-1075","DOIUrl":"10.21037/tlcr-2025-1075","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"5198-5199"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683402/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715807","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: Systemic inflammation may reduce the efficacy of immunotherapy. For advanced non-small cell lung cancer (NSCLC) with programmed death-ligand 1 (PD-L1) expression ≥50% and without driver gene mutations, both immunotherapy monotherapy and combination chemoimmunotherapy are considered standard treatment options; however, it remains unclear which treatment is more appropriate depending on the degree of inflammation. This study investigated the impact of inflammation on the effectiveness of pembrolizumab monotherapy versus combination chemoimmunotherapy.
Methods: In this retrospective multicenter cohort study, we included patients with advanced NSCLC from 13 Japanese institutions who received pembrolizumab monotherapy or combination chemoimmunotherapy as first-line treatment between March 2017 and October 2021. The systemic immune-inflammation index (SII) was used as an inflammation marker, with a cutoff value of 1,444 based on previous data. Patients were classified into high (SII ≥1,444) and low (SII <1,444) inflammation groups.
Results: This study included 347 patients with advanced NSCLC: 212 (61.1%) in the pembrolizumab group and 135 (38.9%) in the combination chemoimmunotherapy group. In the high inflammation population, there were no significant differences in progression-free survival (PFS) and overall survival (OS) between the pembrolizumab and combination chemoimmunotherapy groups. In the low inflammation population, PFS and OS were significantly improved in the combination chemoimmunotherapy group compared to the pembrolizumab group [median PFS: 8.8 vs. 16.0 months, hazard ratio (HR) =0.69, P=0.04; median OS: 29.4 vs. not reached (NR), HR =0.55, P=0.007].
Conclusions: In patients with advanced, driver gene mutation-negative NSCLC, high PD-L1 expression (≥50%), and low systemic inflammation, combination chemoimmunotherapy significantly improved PFS and OS compared to pembrolizumab monotherapy.
{"title":"Selecting optimal immunotherapy based on inflammation in stage IV PD-L1 ≥50% gene mutation-negative non-small cell lung cancer: pembrolizumab monotherapy versus combination chemoimmunotherapy.","authors":"Yusuke Kunimatsu, Naoya Nishioka, Tadaaki Yamada, Hayato Kawachi, Motohiro Tamiya, Yoshiki Negi, Yasuhiro Goto, Akira Nakao, Shinsuke Shiotsu, Keiko Tanimura, Takayuki Takeda, Asuka Okada, Taishi Harada, Koji Date, Yusuke Chihara, Isao Hasegawa, Nobuyo Tamiya, Masaki Ishida, Takashi Kijima, Koichi Takayama","doi":"10.21037/tlcr-2025-873","DOIUrl":"10.21037/tlcr-2025-873","url":null,"abstract":"<p><strong>Background: </strong>Systemic inflammation may reduce the efficacy of immunotherapy. For advanced non-small cell lung cancer (NSCLC) with programmed death-ligand 1 (PD-L1) expression ≥50% and without driver gene mutations, both immunotherapy monotherapy and combination chemoimmunotherapy are considered standard treatment options; however, it remains unclear which treatment is more appropriate depending on the degree of inflammation. This study investigated the impact of inflammation on the effectiveness of pembrolizumab monotherapy versus combination chemoimmunotherapy.</p><p><strong>Methods: </strong>In this retrospective multicenter cohort study, we included patients with advanced NSCLC from 13 Japanese institutions who received pembrolizumab monotherapy or combination chemoimmunotherapy as first-line treatment between March 2017 and October 2021. The systemic immune-inflammation index (SII) was used as an inflammation marker, with a cutoff value of 1,444 based on previous data. Patients were classified into high (SII ≥1,444) and low (SII <1,444) inflammation groups.</p><p><strong>Results: </strong>This study included 347 patients with advanced NSCLC: 212 (61.1%) in the pembrolizumab group and 135 (38.9%) in the combination chemoimmunotherapy group. In the high inflammation population, there were no significant differences in progression-free survival (PFS) and overall survival (OS) between the pembrolizumab and combination chemoimmunotherapy groups. In the low inflammation population, PFS and OS were significantly improved in the combination chemoimmunotherapy group compared to the pembrolizumab group [median PFS: 8.8 <i>vs.</i> 16.0 months, hazard ratio (HR) =0.69, P=0.04; median OS: 29.4 <i>vs.</i> not reached (NR), HR =0.55, P=0.007].</p><p><strong>Conclusions: </strong>In patients with advanced, driver gene mutation-negative NSCLC, high PD-L1 expression (≥50%), and low systemic inflammation, combination chemoimmunotherapy significantly improved PFS and OS compared to pembrolizumab monotherapy.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":"14 11","pages":"4733-4745"},"PeriodicalIF":3.5,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12683408/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145715819","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}