Objectives: To investigate the association between CT attenuation and zero echo time (ZTE) MRI detectability of subsolid pulmonary nodules (SSNs), and to identify a potential attenuation threshold to inform radiation-free follow-up approaches.
Materials and methods: Patients with SSNs ≤ 2 cm scheduled for surgical resection were prospectively enrolled between July 2022 and June 2023. Two radiologists reviewed nodule type (part-solid nodule [PSN] and ground-glass nodule [GGN]), size, location, and attenuation on preoperative CT, and evaluated detectability on ZTE-MRI. Associations between CT features, pathology, and ZTE-MRI detectability were analyzed using univariable and multivariable methods, and predictive performance was assessed with ROC curve analysis.
Results: Thirty-nine patients with 64 SSNs (median diameter, 6.6 mm) participated in the study. Of these, 43 (67.2%) SSNs were identified on ZTE-MRI, including 11 PSNs (100%) and 32 GGNs (60.4%). CT attenuation was the most significant predictor for ZTE-MRI detectability (AUC, 0.993; P < .001), with an optimal threshold of -637.6 HU (sensitivity, 95.2%; specificity, 100.0%). Interreader agreement for ZTE-MRI measurements was high (intraclass correlation coefficient [ICC], 0.986). The size measurements between CT and ZTE-MRI demonstrated minimal bias, up to 0.3 mm. Surgical resection and pathological confirmation were performed for 47 SSNs. Detectable nodules were more frequently invasive adenocarcinomas (IACs) than undetectable nodules (94.1%, 32/34 vs. 30.8%, 4/13; P < .001).
Conclusion: ZTE-MRI detectability of SSNs was significantly associated with CT attenuation, with a potential threshold of -637.6 HU, and correlated with pathological invasiveness. These results may contribute to individualized follow-up protocols for SSNs initially identified by CT imaging.
{"title":"The Impact of CT Attenuation on Subsolid Pulmonary Nodule Detection With the Zero Echo Time MRI Technique.","authors":"Wan-Ting Tao, Tsai-Wang Huang, Hsian-He Hsu, Shih-Wei Chiang, Hsu-Kai Huang, Wen-Chiuan Tsai, Kai-Hsiung Ko","doi":"10.1016/j.cllc.2025.10.006","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.10.006","url":null,"abstract":"<p><strong>Objectives: </strong>To investigate the association between CT attenuation and zero echo time (ZTE) MRI detectability of subsolid pulmonary nodules (SSNs), and to identify a potential attenuation threshold to inform radiation-free follow-up approaches.</p><p><strong>Materials and methods: </strong>Patients with SSNs ≤ 2 cm scheduled for surgical resection were prospectively enrolled between July 2022 and June 2023. Two radiologists reviewed nodule type (part-solid nodule [PSN] and ground-glass nodule [GGN]), size, location, and attenuation on preoperative CT, and evaluated detectability on ZTE-MRI. Associations between CT features, pathology, and ZTE-MRI detectability were analyzed using univariable and multivariable methods, and predictive performance was assessed with ROC curve analysis.</p><p><strong>Results: </strong>Thirty-nine patients with 64 SSNs (median diameter, 6.6 mm) participated in the study. Of these, 43 (67.2%) SSNs were identified on ZTE-MRI, including 11 PSNs (100%) and 32 GGNs (60.4%). CT attenuation was the most significant predictor for ZTE-MRI detectability (AUC, 0.993; P < .001), with an optimal threshold of -637.6 HU (sensitivity, 95.2%; specificity, 100.0%). Interreader agreement for ZTE-MRI measurements was high (intraclass correlation coefficient [ICC], 0.986). The size measurements between CT and ZTE-MRI demonstrated minimal bias, up to 0.3 mm. Surgical resection and pathological confirmation were performed for 47 SSNs. Detectable nodules were more frequently invasive adenocarcinomas (IACs) than undetectable nodules (94.1%, 32/34 vs. 30.8%, 4/13; P < .001).</p><p><strong>Conclusion: </strong>ZTE-MRI detectability of SSNs was significantly associated with CT attenuation, with a potential threshold of -637.6 HU, and correlated with pathological invasiveness. These results may contribute to individualized follow-up protocols for SSNs initially identified by CT imaging.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145457875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-10DOI: 10.1016/j.cllc.2025.10.004
Masashi Azuma, Alexander Nguyen, Anastasiia K Tompkins, Kristine Chin, Bradley Walker, Rishabh Matta, Daohai Yu, Cherie P Erkmen
Background: Lung cancer screening (LCS) with Low-Dose Computed Tomography (LDCT) significantly reduces cancer mortality but remains substantially underutilized. This study examines how social determinants of health (SDOH), including financial concerns, transportation barriers, and patient-provider trust, influence adherence to annual LCS.
Methods: A prospective cohort study was conducted at an urban, safety-net academic health system. Participants who initially underwent LDCT but failed to adhere to annual follow-up were surveyed, assessing demographics, financial and transportation concerns, patient-provider relationships, and experiences of racial discrimination. Data was analyzed using descriptive statistics, chi-square tests, and Spearman correlations.
Results: Among surveyors (n = 236), cost concerns (54.4%) and access to transportation (13.3%) were significant barriers to LCS adherence. Access to transportation was more limited for non-White (22.2%) and female (18.4%) populations, compared to White (6.5%, P < .01) and male (7.1%, P = .02) populations. However, there was no difference in cost concerns (P = .50, P = .89). Additionally, non-White (4.51/5 vs. 4.26/5, P < .01) and female (4.44/5 vs. 4.31/5, P = .04) patients reported higher levels of trust and comfort with providers comparing to their counter cohorts. Perceived racial discrimination in healthcare remained higher in non-White participants (1.53/5 vs. 1.25/5, P < .01) without effect in gender cohorts (P = .24).
Conclusions: LCS can decrease the chance of lung cancer death among eligible individuals but is drastically underutilized. Most people not adhering to LCS cited cost concerns, despite coverage by insurers. Patient-provider trust can be leveraged to assess and address individual barriers to LCS.
背景:肺癌筛查(LCS)低剂量计算机断层扫描(LDCT)可显著降低癌症死亡率,但仍未得到充分利用。本研究考察了健康的社会决定因素(SDOH),包括财务问题、交通障碍和患者-提供者信任,如何影响遵守年度LCS。方法:前瞻性队列研究在一个城市,安全网学术卫生系统进行。最初接受LDCT但未能坚持每年随访的参与者进行了调查,评估人口统计学,财务和交通问题,患者-提供者关系和种族歧视经历。数据分析采用描述性统计、卡方检验和Spearman相关性。结果:在调查人员(n = 236)中,成本问题(54.4%)和交通运输(13.3%)是遵守LCS的重大障碍。与白人(6.5%,P < 0.01)和男性(7.1%,P = 0.02)人群相比,非白人(22.2%)和女性(18.4%)人群的交通可及性更有限。然而,在成本问题上没有差异(P = 0.50, P = 0.89)。此外,非白人(4.51/5 vs. 4.26/5, P < 0.01)和女性(4.44/5 vs. 4.31/5, P = 0.04)患者报告对提供者的信任度和舒适度高于对照组。在非白人参与者中,医疗保健中的种族歧视感知仍然较高(1.53/5比1.25/5,P < 0.01),而在性别队列中没有影响(P = 0.24)。结论:LCS可以降低符合条件的个体的肺癌死亡机会,但未得到充分利用。大多数不遵守LCS的人提到了成本问题,尽管保险公司提供了保险。可以利用患者-提供者信任来评估和解决LCS的个别障碍。
{"title":"Lost to Follow-up: Social Determinants and Patient Perceptions in Lung Cancer Screening Adherence.","authors":"Masashi Azuma, Alexander Nguyen, Anastasiia K Tompkins, Kristine Chin, Bradley Walker, Rishabh Matta, Daohai Yu, Cherie P Erkmen","doi":"10.1016/j.cllc.2025.10.004","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.10.004","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer screening (LCS) with Low-Dose Computed Tomography (LDCT) significantly reduces cancer mortality but remains substantially underutilized. This study examines how social determinants of health (SDOH), including financial concerns, transportation barriers, and patient-provider trust, influence adherence to annual LCS.</p><p><strong>Methods: </strong>A prospective cohort study was conducted at an urban, safety-net academic health system. Participants who initially underwent LDCT but failed to adhere to annual follow-up were surveyed, assessing demographics, financial and transportation concerns, patient-provider relationships, and experiences of racial discrimination. Data was analyzed using descriptive statistics, chi-square tests, and Spearman correlations.</p><p><strong>Results: </strong>Among surveyors (n = 236), cost concerns (54.4%) and access to transportation (13.3%) were significant barriers to LCS adherence. Access to transportation was more limited for non-White (22.2%) and female (18.4%) populations, compared to White (6.5%, P < .01) and male (7.1%, P = .02) populations. However, there was no difference in cost concerns (P = .50, P = .89). Additionally, non-White (4.51/5 vs. 4.26/5, P < .01) and female (4.44/5 vs. 4.31/5, P = .04) patients reported higher levels of trust and comfort with providers comparing to their counter cohorts. Perceived racial discrimination in healthcare remained higher in non-White participants (1.53/5 vs. 1.25/5, P < .01) without effect in gender cohorts (P = .24).</p><p><strong>Conclusions: </strong>LCS can decrease the chance of lung cancer death among eligible individuals but is drastically underutilized. Most people not adhering to LCS cited cost concerns, despite coverage by insurers. Patient-provider trust can be leveraged to assess and address individual barriers to LCS.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145444111","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-10DOI: 10.1016/j.cllc.2025.10.008
Duy Pham, Nikita Thakur, Ju Ae Park, Wei Nie, Michael Correa, Hongkun Wang, Melanie Subramanian, Michael Weyant, Christopher B Johnson, Kei Suzuki
Objective: There is a paucity of pretreatment, noninvasive assessment of tumor characteristics in non-small-cell lung cancer (NSCLC) undergoing stereotactic body radiation therapy (SBRT). Our goal was to determine if Computer Assisted Nodule Analysis and Risk Yield (CANARY) could stratify risk for recurrence in patients undergoing SBRT for clinical stage I NSCLC.
Methods: We performed a retrospective review of NSCLC patients who underwent SBRT from 2016 to 2022. Recurrence dates were collected from the date of therapy to May 2023. Pretreatment imaging was entered into the CANARY software, the correct lesion verified and demarcated, and each lesion was categorized into good, intermediate, and poor Score Indicative of Lung Cancer Aggression (SILA). Kaplan-Meier methodology was used to analyze the recurrence free survival (RFS), and Log-rank test was used for group comparison.
Results: The SBRT cohort included 85 patients. By clinical stage, there were 4 (4.7%) IA1, 41 (48.2%) IA2, 30 (35.3%) IA3, and 10 (11.8%) IB. By histology, 48 (56.5%) patients were adenocarcinoma, 23 (27.1%) were squamous cell carcinoma, and 14 (16.5%) were NSCLC. The 2-year RFS was 68.5%. By CANARY, 11 (12.9%) patients were considered to have good risk, 14 (17.6%) intermediate risk, and 60 (70.6%) poor risk by their SILA scores. Their 2-year RFS was 100.0% (100.0%-100.0%), 80.2% (58.7%-100.0%), and 68.1% (54.4%-85.3%), respectively. Intermediate and poor SILA scores had worse RFS than good SILA score patients (P = .09).
Conclusions: CANARY can potentially risk stratify recurrence in clinical stage I NSCLC patients undergoing SBRT.
{"title":"Computer Assisted Nodule Analysis and Risk Yield Outcomes May be Associated with Recurrence after Stereotactic Body Radiation Therapy in Clinical Stage I Non-Small-Cell Lung Cancer.","authors":"Duy Pham, Nikita Thakur, Ju Ae Park, Wei Nie, Michael Correa, Hongkun Wang, Melanie Subramanian, Michael Weyant, Christopher B Johnson, Kei Suzuki","doi":"10.1016/j.cllc.2025.10.008","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.10.008","url":null,"abstract":"<p><strong>Objective: </strong>There is a paucity of pretreatment, noninvasive assessment of tumor characteristics in non-small-cell lung cancer (NSCLC) undergoing stereotactic body radiation therapy (SBRT). Our goal was to determine if Computer Assisted Nodule Analysis and Risk Yield (CANARY) could stratify risk for recurrence in patients undergoing SBRT for clinical stage I NSCLC.</p><p><strong>Methods: </strong>We performed a retrospective review of NSCLC patients who underwent SBRT from 2016 to 2022. Recurrence dates were collected from the date of therapy to May 2023. Pretreatment imaging was entered into the CANARY software, the correct lesion verified and demarcated, and each lesion was categorized into good, intermediate, and poor Score Indicative of Lung Cancer Aggression (SILA). Kaplan-Meier methodology was used to analyze the recurrence free survival (RFS), and Log-rank test was used for group comparison.</p><p><strong>Results: </strong>The SBRT cohort included 85 patients. By clinical stage, there were 4 (4.7%) IA1, 41 (48.2%) IA2, 30 (35.3%) IA3, and 10 (11.8%) IB. By histology, 48 (56.5%) patients were adenocarcinoma, 23 (27.1%) were squamous cell carcinoma, and 14 (16.5%) were NSCLC. The 2-year RFS was 68.5%. By CANARY, 11 (12.9%) patients were considered to have good risk, 14 (17.6%) intermediate risk, and 60 (70.6%) poor risk by their SILA scores. Their 2-year RFS was 100.0% (100.0%-100.0%), 80.2% (58.7%-100.0%), and 68.1% (54.4%-85.3%), respectively. Intermediate and poor SILA scores had worse RFS than good SILA score patients (P = .09).</p><p><strong>Conclusions: </strong>CANARY can potentially risk stratify recurrence in clinical stage I NSCLC patients undergoing SBRT.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145444169","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: Occult lymph node (LN) metastasis, indicating pathological LN involvement, is often observed in clinical N0 (cN0) early-stage non-small cell lung cancer (NSCLC). Identifying preoperative predictors of occult LN metastasis (pathologically N1 and N2) is crucial for determining the surgical procedure for cN0 NSCLC. This study aimed to investigate the role of genetic mutations in predicting occult LN metastasis and their influence on surgical strategies.
Materials and methods: We retrospectively reviewed patients who underwent lobectomy or segmentectomy for cN0 NSCLC with pathological stages higher than IB, between May 2017 and April 2024. Clinicopathological characteristics and genetic mutations were analyzed. Occult LN metastasis was not detected by imaging but identified through histopathology.
Results: We evaluated 644 patients, median age 71 years. Occult LN metastases were observed in 179 (27.8%) patients. EGFR mutations and ALK rearrangements were significantly associated with occult LN metastasis (EGFR, P = .04; ALK, P = .007), particularly EGFR exon 19 deletions (P = .006). Multivariate analysis confirmed these as significant predictors (P = .006). Notably, patients with these mutations had longer recurrence-free survival (RFS) than those without (HR 1.27, 95% CI, 0.90-1.80, P = .02). For patients with EGFR mutations and ALK rearrangements, RFS was significantly shorter after segmentectomy (HR 3.18, 95% CI, 1.02-9.99, P = .04) and longer after lobectomy (HR 1.28, 95% CI, 0.87-1.90, P = .01).
Conclusion: Genomic profiles, such as EGFR mutations and ALK rearrangements, are associated with occult LN metastasis and outcomes in cN0 NSCLC. Integrating genetic assessments into preoperative planning may optimize surgical strategies and improve prognosis.
目的:隐匿淋巴结(LN)转移是临床N0 (cN0)早期非小细胞肺癌(NSCLC)中常见的病理淋巴结转移。确定隐匿性淋巴结转移的术前预测因子(病理上的N1和N2)对于确定cN0型非小细胞肺癌的手术方法至关重要。本研究旨在探讨基因突变在预测隐匿性淋巴结转移中的作用及其对手术策略的影响。材料和方法:我们回顾性分析了2017年5月至2024年4月期间接受病理分期高于IB的cN0 NSCLC肺叶切除术或节段切除术的患者。分析临床病理特征及基因突变。隐匿性淋巴结转移未通过影像学发现,但通过组织病理学发现。结果:我们评估了644例患者,中位年龄71岁。179例(27.8%)患者出现隐匿性淋巴结转移。EGFR突变和ALK重排与隐匿性淋巴结转移显著相关(EGFR, P = 0.04; ALK, P = 0.007),尤其是EGFR外显子19缺失(P = 0.006)。多变量分析证实这些是显著的预测因子(P = .006)。值得注意的是,有这些突变的患者比没有突变的患者有更长的无复发生存期(RFS) (HR 1.27, 95% CI, 0.90-1.80, P = 0.02)。对于EGFR突变和ALK重排的患者,节段切除术后RFS显著缩短(HR 3.18, 95% CI, 1.02-9.99, P = 0.04),肺叶切除术后RFS显著延长(HR 1.28, 95% CI, 0.87-1.90, P = 0.01)。结论:基因组谱,如EGFR突变和ALK重排,与cN0 NSCLC的隐匿性LN转移和预后相关。将基因评估纳入术前计划可以优化手术策略并改善预后。
{"title":"Genomic Profiles as Predictors of Occult Lymph Node Metastasis and Clinical Outcomes in Early-Stage Clinical N0 Non-Small Cell Lung Cancer.","authors":"Chihiro Takemura, Tatsuya Yoshida, Yukihiro Yoshida, Ryoko Inaba Higashiyama, Hidehito Horinouchi, Hiroshi Igaki, Noboru Yamamoto, Yuichiro Ohe, Yasushi Yatabe, Shun-Ichi Watanabe","doi":"10.1016/j.cllc.2025.10.002","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.10.002","url":null,"abstract":"<p><strong>Objective: </strong>Occult lymph node (LN) metastasis, indicating pathological LN involvement, is often observed in clinical N0 (cN0) early-stage non-small cell lung cancer (NSCLC). Identifying preoperative predictors of occult LN metastasis (pathologically N1 and N2) is crucial for determining the surgical procedure for cN0 NSCLC. This study aimed to investigate the role of genetic mutations in predicting occult LN metastasis and their influence on surgical strategies.</p><p><strong>Materials and methods: </strong>We retrospectively reviewed patients who underwent lobectomy or segmentectomy for cN0 NSCLC with pathological stages higher than IB, between May 2017 and April 2024. Clinicopathological characteristics and genetic mutations were analyzed. Occult LN metastasis was not detected by imaging but identified through histopathology.</p><p><strong>Results: </strong>We evaluated 644 patients, median age 71 years. Occult LN metastases were observed in 179 (27.8%) patients. EGFR mutations and ALK rearrangements were significantly associated with occult LN metastasis (EGFR, P = .04; ALK, P = .007), particularly EGFR exon 19 deletions (P = .006). Multivariate analysis confirmed these as significant predictors (P = .006). Notably, patients with these mutations had longer recurrence-free survival (RFS) than those without (HR 1.27, 95% CI, 0.90-1.80, P = .02). For patients with EGFR mutations and ALK rearrangements, RFS was significantly shorter after segmentectomy (HR 3.18, 95% CI, 1.02-9.99, P = .04) and longer after lobectomy (HR 1.28, 95% CI, 0.87-1.90, P = .01).</p><p><strong>Conclusion: </strong>Genomic profiles, such as EGFR mutations and ALK rearrangements, are associated with occult LN metastasis and outcomes in cN0 NSCLC. Integrating genetic assessments into preoperative planning may optimize surgical strategies and improve prognosis.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145430551","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-06DOI: 10.1016/j.cllc.2025.10.001
Paul K Paik, Jianjun Zhang, Howard Jack West, Jonathan W Riess
Although the pharmacologic management of non-small cell lung cancer (NSCLC) has advanced substantially in the past 2 decades, disparities in the applicability to different histologic subtypes remain. Several treatment options are not suitable for squamous NSCLC, with use restricted to nonsquamous NSCLC (eg, bevacizumab and pemetrexed) because of safety concerns or comparative activity. Differences in mutational landscapes and a lack of matched targeted therapies specific to squamous NSCLC oncogenic aberrations present additional limitations. In the absence of suitable targeted therapies, immunotherapy with or without chemotherapy is the mainstay of squamous NSCLC treatment; however, survival-related outcomes remain poorer for patients with squamous than with nonsquamous NSCLC. Several studies are investigating promising drug therapies for patients with squamous NSCLC. This review seeks to summarize the current and emerging treatment options for patients with squamous NSCLC.
{"title":"Squamous Non-Small Cell Lung Cancer: Current and Emerging Treatment Options.","authors":"Paul K Paik, Jianjun Zhang, Howard Jack West, Jonathan W Riess","doi":"10.1016/j.cllc.2025.10.001","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.10.001","url":null,"abstract":"<p><p>Although the pharmacologic management of non-small cell lung cancer (NSCLC) has advanced substantially in the past 2 decades, disparities in the applicability to different histologic subtypes remain. Several treatment options are not suitable for squamous NSCLC, with use restricted to nonsquamous NSCLC (eg, bevacizumab and pemetrexed) because of safety concerns or comparative activity. Differences in mutational landscapes and a lack of matched targeted therapies specific to squamous NSCLC oncogenic aberrations present additional limitations. In the absence of suitable targeted therapies, immunotherapy with or without chemotherapy is the mainstay of squamous NSCLC treatment; however, survival-related outcomes remain poorer for patients with squamous than with nonsquamous NSCLC. Several studies are investigating promising drug therapies for patients with squamous NSCLC. This review seeks to summarize the current and emerging treatment options for patients with squamous NSCLC.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145548568","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-03DOI: 10.1016/j.cllc.2025.09.002
Shirish M Gadgeel, Misako Nagasaka, Karen Dziubek, Thomas Braun, Khaled Hassan, Haiying Cheng, Balazs Halmos, Antoinette Wozniak, James Stevenson, Pradnya Patil, Nathan Pennell, Mary Jo Fidler, Angel Qin, Zeqi Niu, Sunitha Nagrath, Gregory P Kalemkerian
Introduction: Immune checkpoint inhibitors have limited efficacy in patients with EGFR-mutant (EGFR+) and ALK-rearranged (ALK+) non-small cell lung cancer (NSCLC). We conducted a phase II study to evaluate the efficacy of pembrolizumab with carboplatin and pemetrexed in these patients.
Patients and methods: EGFR+ or ALK+ NSCLC patients, previously treated with targeted therapy, were eligible. Carboplatin, pemetrexed and pembrolizumab were administered every 3 weeks for 4 cycles followed by maintenance pemetrexed and pembrolizumab. The primary endpoint was response rate (RR). Blood for circulating tumor cells (CTCs) was collected prior to the 1st and 3rd cycles. The plan was to enroll 28 evaluable patients in both EGFR+ and ALK+ cohorts.
Results: Of the 33 patients enrolled, 26 had EGFR+ and 7 had ALK+ NSCLC. RR (95% CI,) was 46% (27%, 67%) in EGFR+ and 29% (4%, 71%), in ALK+ patients, respectively. Median progression free survival (PFS) and overall survival (OS) in the EGFR+ cohort were 8.3 months (7.2-16.5) and 22.2 months (20.6-NE), respectively. In the ALK+ cohort, median PFS and OS were both 2.9 months. The median CTC count at baseline in 15 evaluable EGFR+ patients was 4 cells/mL (0-23). OS among EGFR+ patients with decreasing vs. increasing CTC count during treatment was not reached vs. 18.5 months, respectively (P = .52). The most common adverse events were fatigue, nausea, anemia and AST/ALT elevation.
Conclusion: Pembrolizumab in combination with chemotherapy demonstrated encouraging RR of 42% and OS of 22 months among patients with recurrent EGFR+ NSCLC. The efficacy in ALK+ patients was not encouraging.
{"title":"Pembrolizumab in Combination With Platinum-Based Chemotherapy in Patients With Recurrent EGFR and ALK Gene Altered Non-Small-Cell Lung Cancer (NSCLC).","authors":"Shirish M Gadgeel, Misako Nagasaka, Karen Dziubek, Thomas Braun, Khaled Hassan, Haiying Cheng, Balazs Halmos, Antoinette Wozniak, James Stevenson, Pradnya Patil, Nathan Pennell, Mary Jo Fidler, Angel Qin, Zeqi Niu, Sunitha Nagrath, Gregory P Kalemkerian","doi":"10.1016/j.cllc.2025.09.002","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.09.002","url":null,"abstract":"<p><strong>Introduction: </strong>Immune checkpoint inhibitors have limited efficacy in patients with EGFR-mutant (EGFR+) and ALK-rearranged (ALK+) non-small cell lung cancer (NSCLC). We conducted a phase II study to evaluate the efficacy of pembrolizumab with carboplatin and pemetrexed in these patients.</p><p><strong>Patients and methods: </strong>EGFR+ or ALK+ NSCLC patients, previously treated with targeted therapy, were eligible. Carboplatin, pemetrexed and pembrolizumab were administered every 3 weeks for 4 cycles followed by maintenance pemetrexed and pembrolizumab. The primary endpoint was response rate (RR). Blood for circulating tumor cells (CTCs) was collected prior to the 1st and 3rd cycles. The plan was to enroll 28 evaluable patients in both EGFR+ and ALK+ cohorts.</p><p><strong>Results: </strong>Of the 33 patients enrolled, 26 had EGFR+ and 7 had ALK+ NSCLC. RR (95% CI,) was 46% (27%, 67%) in EGFR+ and 29% (4%, 71%), in ALK+ patients, respectively. Median progression free survival (PFS) and overall survival (OS) in the EGFR+ cohort were 8.3 months (7.2-16.5) and 22.2 months (20.6-NE), respectively. In the ALK+ cohort, median PFS and OS were both 2.9 months. The median CTC count at baseline in 15 evaluable EGFR+ patients was 4 cells/mL (0-23). OS among EGFR+ patients with decreasing vs. increasing CTC count during treatment was not reached vs. 18.5 months, respectively (P = .52). The most common adverse events were fatigue, nausea, anemia and AST/ALT elevation.</p><p><strong>Conclusion: </strong>Pembrolizumab in combination with chemotherapy demonstrated encouraging RR of 42% and OS of 22 months among patients with recurrent EGFR+ NSCLC. The efficacy in ALK+ patients was not encouraging.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145228514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-03DOI: 10.1016/j.cllc.2025.09.015
Ilaria Attili, Gloria Pellizzari, Luca Bertolaccini, Carla Corvaja, Davide Vacirca, Mariano Lombardi, Gianluca Spitaleri, Pamela Trillo Aliaga, Ester Del Signore, Antonio Passaro, Juliana Guarize, Elena Guerini-Rocco, Nicola Fusco, Lorenzo Spaggiari, Filippo de Marinis
Introduction: In resected non-small cell lung cancer (NSCLC), molecular testing is currently limited to EGFR mutations and ALK rearrangements, as these guide approved adjuvant therapies. The role of next-generation sequencing (NGS), routinely used in metastatic NSCLC, remains unclear in earlier stages. The prevalence of other driver mutations and their impact on outcomes is not well established.
Methods: We retrospectively analyzed clinical, molecular, and survival data from patients (pts) with stage IA-IIIB NSCLC (AJCC 8th) who underwent surgery and NGS at our Institute from January 2020 to December 2023. The primary endpoint was the prevalence of driver alterations. Exploratory analyses assessed recurrence, disease-free survival (DFS), overall survival (OS), and correlation with mutation status.
Results: Of 221 pts, 216 were eligible. Oncogenic alterations were found in 71% (73% in stage I), most commonly KRAS (30%) and EGFR (26%), followed by MET exon 14 skipping (6%), BRAF (4%), HER2 exon 20 mutations (3%), and ALK and RET rearrangements (1%). Alteration distribution differed by sex and smoking status. With a median follow-up of 20 months, 36% of pts experienced recurrence, including 10 stage I cases. Median time to recurrence was 14 months. Recurrence rates were higher in pts with driver mutant NSCLC (39.6%) versus wild-type (29.6%). Highest recurrence was seen in pts with oncogenic fusion positive NSCLC and EGFR exon 20 insertions.
Conclusions: Driver mutations were detected in 70% of resected NSCLC, including stage I. The recurrence patterns observed support integrating NGS into early-stage management and exploring tailored adjuvant therapies, even for stage I tumors.
{"title":"Actionable Gene Alterations in Resected Non-Small Cell Lung Cancer: Primary Results From the AGA-R Study.","authors":"Ilaria Attili, Gloria Pellizzari, Luca Bertolaccini, Carla Corvaja, Davide Vacirca, Mariano Lombardi, Gianluca Spitaleri, Pamela Trillo Aliaga, Ester Del Signore, Antonio Passaro, Juliana Guarize, Elena Guerini-Rocco, Nicola Fusco, Lorenzo Spaggiari, Filippo de Marinis","doi":"10.1016/j.cllc.2025.09.015","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.09.015","url":null,"abstract":"<p><strong>Introduction: </strong>In resected non-small cell lung cancer (NSCLC), molecular testing is currently limited to EGFR mutations and ALK rearrangements, as these guide approved adjuvant therapies. The role of next-generation sequencing (NGS), routinely used in metastatic NSCLC, remains unclear in earlier stages. The prevalence of other driver mutations and their impact on outcomes is not well established.</p><p><strong>Methods: </strong>We retrospectively analyzed clinical, molecular, and survival data from patients (pts) with stage IA-IIIB NSCLC (AJCC 8th) who underwent surgery and NGS at our Institute from January 2020 to December 2023. The primary endpoint was the prevalence of driver alterations. Exploratory analyses assessed recurrence, disease-free survival (DFS), overall survival (OS), and correlation with mutation status.</p><p><strong>Results: </strong>Of 221 pts, 216 were eligible. Oncogenic alterations were found in 71% (73% in stage I), most commonly KRAS (30%) and EGFR (26%), followed by MET exon 14 skipping (6%), BRAF (4%), HER2 exon 20 mutations (3%), and ALK and RET rearrangements (1%). Alteration distribution differed by sex and smoking status. With a median follow-up of 20 months, 36% of pts experienced recurrence, including 10 stage I cases. Median time to recurrence was 14 months. Recurrence rates were higher in pts with driver mutant NSCLC (39.6%) versus wild-type (29.6%). Highest recurrence was seen in pts with oncogenic fusion positive NSCLC and EGFR exon 20 insertions.</p><p><strong>Conclusions: </strong>Driver mutations were detected in 70% of resected NSCLC, including stage I. The recurrence patterns observed support integrating NGS into early-stage management and exploring tailored adjuvant therapies, even for stage I tumors.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421432","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-02DOI: 10.1016/j.cllc.2025.09.014
Elisa Daffré, Samir Bouam, Salvatore Strano, Kim Blanc, Audrey Lupo-Mansuet, Emelyne Canny, Ludovic Fournel, Antonio Bobbio, Mathilde Prieto, Marco Alifano
Objectives: Lung cancer may require complex interventions such as pneumonectomy. This study investigates late survival after pneumonectomy and assesses the need for late rehospitalizations.
Methods: We performed a retrospective analysis of 479 consecutive patients with lung cancer undergoing pneumonectomy at our institution between 2005 and 2015. Survival was assessed at 3, 5, and 8 years and Cox multivariate analysis was used to identify independent predictors. The National anonymized medico-economic database (PMSI) was used to study early and late (up to 10 years after surgery) postpneumonectomy hospital trajectories (excluding routine follow-up visits) of patients in the last 2 years of the study period (n = 179).
Results: The mean age of patients was 62.7 ± 9.9 years and 74.9% were men; 56.18% had FEV1 below 80% of predicted. Stage III accounted for 62.84% of patients. Overall survival at 3, 5 and 8 years was 58.9%, 47.6% and 39.6%, respectively. Male sex, active tobacco smoking, Charlson Comorbidity Index >5, stage III-IV, and occurrence of Clavien Dindo≥3 postoperative complications were independent predictors of worse survival. After pneumonectomy, a total of 4845 hospitalisations were recorded in 179 patients, 886 for postoperative adjuvant treatments. Forty-five out of the 179 patients had a late hospitalisation for lung cancer recurrence, with an average delay of 23.3 months. There were 25 readmissions, involving 20/179 patients, for respiratory failure, whereas 15/179 patients were readmitted for cardiac failure. In addition, 9/179 patients were readmitted for a new nonpulmonary cancer.
Conclusions: Despite the strong physiological impact, pneumonectomy has satisfactory short and long-term results.
{"title":"Late Survival and Hospital Trajectory After Pneumonectomy for Non-small Cell Lung Cancer.","authors":"Elisa Daffré, Samir Bouam, Salvatore Strano, Kim Blanc, Audrey Lupo-Mansuet, Emelyne Canny, Ludovic Fournel, Antonio Bobbio, Mathilde Prieto, Marco Alifano","doi":"10.1016/j.cllc.2025.09.014","DOIUrl":"https://doi.org/10.1016/j.cllc.2025.09.014","url":null,"abstract":"<p><strong>Objectives: </strong>Lung cancer may require complex interventions such as pneumonectomy. This study investigates late survival after pneumonectomy and assesses the need for late rehospitalizations.</p><p><strong>Methods: </strong>We performed a retrospective analysis of 479 consecutive patients with lung cancer undergoing pneumonectomy at our institution between 2005 and 2015. Survival was assessed at 3, 5, and 8 years and Cox multivariate analysis was used to identify independent predictors. The National anonymized medico-economic database (PMSI) was used to study early and late (up to 10 years after surgery) postpneumonectomy hospital trajectories (excluding routine follow-up visits) of patients in the last 2 years of the study period (n = 179).</p><p><strong>Results: </strong>The mean age of patients was 62.7 ± 9.9 years and 74.9% were men; 56.18% had FEV1 below 80% of predicted. Stage III accounted for 62.84% of patients. Overall survival at 3, 5 and 8 years was 58.9%, 47.6% and 39.6%, respectively. Male sex, active tobacco smoking, Charlson Comorbidity Index >5, stage III-IV, and occurrence of Clavien Dindo≥3 postoperative complications were independent predictors of worse survival. After pneumonectomy, a total of 4845 hospitalisations were recorded in 179 patients, 886 for postoperative adjuvant treatments. Forty-five out of the 179 patients had a late hospitalisation for lung cancer recurrence, with an average delay of 23.3 months. There were 25 readmissions, involving 20/179 patients, for respiratory failure, whereas 15/179 patients were readmitted for cardiac failure. In addition, 9/179 patients were readmitted for a new nonpulmonary cancer.</p><p><strong>Conclusions: </strong>Despite the strong physiological impact, pneumonectomy has satisfactory short and long-term results.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-01DOI: 10.1016/j.cllc.2025.09.009
Bob T Li, Leena Gandhi, Vignesh Ravichandran, Benjamin Besse, Julien Mazières, Geoffrey I Shapiro, Valentina Boni, Saiama N Waqar, Haeseong Park, David I Quinn, Alejandro Martinez, Salomon M Stemmer, Alexis B Cortot, Fabrice Barlesi, Elisabeth Quoix, Mark E Burkard, S Duygu Selcuklu, Casey Hunt, Mark T A Donoghue, Mark G Kris, Judith Bebchuk, Lisa D Eli, Leanne McCulloch, David B Solit, Pasi A Jänne
Background: ERBB2 mutations are oncogenic drivers in 2% to 4% of lung cancers and potentially actionable using HER2 tyrosine kinase inhibitors.
Patients and methods: Patients with advanced ERBB2-mutant lung cancer entered 2 phase 2 studies (PUMA-NER-4201 [4201]; PUMA-NER-5201 [SUMMIT]). Patients received neratinib (monotherapy 4201; monotherapy SUMMIT), or neratinib with weekly temsirolimus (combination 4201), or trastuzumab every 3 weeks (combination SUMMIT). Protocol-defined endpoints common to both studies were analyzed. Exploratory genomic analyses were conducted.
Clinicaltrials: gov: NCT01827267; NCT01953926.
Results: Sixty patients in 4201 (neratinib, n = 17; neratinib plus temsirolimus, n = 43) and 78 in SUMMIT (neratinib, n = 26; neratinib plus trastuzumab, n = 52) received study treatment. Objective response rates were 0% (95% confidence interval [CI], 0.0-19.5; 4201) and 3.8% (95% CI, 0.1-19.6; SUMMIT) with neratinib, 14.0% (95% CI, 5.3-27.9) with neratinib plus temsirolimus, and 9.6% (95% CI, 3.2-21.0) with neratinib plus trastuzumab. Five patients whose tumors harbored ERBB2 exon 20 insertion, L755P and D769Y missense mutations, and a novel ERBB2-SHC1 fusion had responses ≥ 1 year (neratinib monotherapy, n = 1; SUMMIT; neratinib plus temsirolimus, n = 2; neratinib plus trastuzumab, n = 2). Grade ≥ 3 treatment-related adverse events occurred in 23.5% (4201) and 34.6% (SUMMIT) of neratinib-treated patients, 37.2% of patients treated with neratinib plus temsirolimus, and 48.1% of patients treated with neratinib plus trastuzumab.
Conclusion: Single-agent neratinib has limited activity in ERBB2-mutated lung cancers. Combinations with temsirolimus or trastuzumab did not markedly improve overall outcomes, producing durable responses in a limited subset of patients.
{"title":"Efficacy of Neratinib-Based Therapy in ERBB2-Mutant Lung Adenocarcinomas: Findings From 2 International Phase 2 Studies.","authors":"Bob T Li, Leena Gandhi, Vignesh Ravichandran, Benjamin Besse, Julien Mazières, Geoffrey I Shapiro, Valentina Boni, Saiama N Waqar, Haeseong Park, David I Quinn, Alejandro Martinez, Salomon M Stemmer, Alexis B Cortot, Fabrice Barlesi, Elisabeth Quoix, Mark E Burkard, S Duygu Selcuklu, Casey Hunt, Mark T A Donoghue, Mark G Kris, Judith Bebchuk, Lisa D Eli, Leanne McCulloch, David B Solit, Pasi A Jänne","doi":"10.1016/j.cllc.2025.09.009","DOIUrl":"10.1016/j.cllc.2025.09.009","url":null,"abstract":"<p><strong>Background: </strong>ERBB2 mutations are oncogenic drivers in 2% to 4% of lung cancers and potentially actionable using HER2 tyrosine kinase inhibitors.</p><p><strong>Patients and methods: </strong>Patients with advanced ERBB2-mutant lung cancer entered 2 phase 2 studies (PUMA-NER-4201 [4201]; PUMA-NER-5201 [SUMMIT]). Patients received neratinib (monotherapy 4201; monotherapy SUMMIT), or neratinib with weekly temsirolimus (combination 4201), or trastuzumab every 3 weeks (combination SUMMIT). Protocol-defined endpoints common to both studies were analyzed. Exploratory genomic analyses were conducted.</p><p><strong>Clinicaltrials: </strong>gov: NCT01827267; NCT01953926.</p><p><strong>Results: </strong>Sixty patients in 4201 (neratinib, n = 17; neratinib plus temsirolimus, n = 43) and 78 in SUMMIT (neratinib, n = 26; neratinib plus trastuzumab, n = 52) received study treatment. Objective response rates were 0% (95% confidence interval [CI], 0.0-19.5; 4201) and 3.8% (95% CI, 0.1-19.6; SUMMIT) with neratinib, 14.0% (95% CI, 5.3-27.9) with neratinib plus temsirolimus, and 9.6% (95% CI, 3.2-21.0) with neratinib plus trastuzumab. Five patients whose tumors harbored ERBB2 exon 20 insertion, L755P and D769Y missense mutations, and a novel ERBB2-SHC1 fusion had responses ≥ 1 year (neratinib monotherapy, n = 1; SUMMIT; neratinib plus temsirolimus, n = 2; neratinib plus trastuzumab, n = 2). Grade ≥ 3 treatment-related adverse events occurred in 23.5% (4201) and 34.6% (SUMMIT) of neratinib-treated patients, 37.2% of patients treated with neratinib plus temsirolimus, and 48.1% of patients treated with neratinib plus trastuzumab.</p><p><strong>Conclusion: </strong>Single-agent neratinib has limited activity in ERBB2-mutated lung cancers. Combinations with temsirolimus or trastuzumab did not markedly improve overall outcomes, producing durable responses in a limited subset of patients.</p>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":" ","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12696912/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145451181","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This study aimed to evaluate whether pretreatment circulating tumor DNA (ctDNA) is a predictive biomarker for recurrence in patients with early-stage non-small cell lung cancer (ES-NSCLC), including those with pathologically unproven (PU) who received ablative radiotherapy.
Materials and methods
Patients clinically diagnosed with cT1-2N0M0 NSCLC treated with radiotherapy alone at a single institute were included. Pretreatment plasma and matched white blood cell samples were subjected to targeted sequencing to detect ctDNA. The association between ctDNA detection and prognosis was evaluated. Integrated analysis including non-squamous cell carcinoma (SQ) cohort and enhancement of recurrence prediction with radiographic features were also evaluated.
Results
Of the 19 patients (10 SQ, and 9 PU) who met the sequence quality criteria, ctDNA was detected in 6. At the median follow-up of 60 months, detectable ctDNA was significantly associated with inferior progression-free survival (PFS) in both the SQ and PU cohorts (P = .01 and .02). In an analysis of 62 patients combined with the non-SQ cohort, ctDNA detection was the only significant factor for PFS. The combination of ctDNA and a high positron emission tomography maximum standardized uptake value provided a better recurrence risk model than ctDNA alone (the C-index improved from 0.63 to 0.72).
Conclusion
These results suggest that pretreatment ctDNA detection can predict recurrence in patients with cT1-2N0M0 NSCLC, including those with PU tumors treated with ablative radiotherapy.
{"title":"Tumor-Agnostic Circulating Tumor DNA Analysis as a Comprehensive Predictive Biomarker of Recurrence after Ablative Radiotherapy for Early-Stage Non-Small Cell Lung Cancer","authors":"Masaki Nakamura , Shun-Ichiro Kageyama , Hidenari Hirata , Masaki Ohira , Shunsuke A. Sakai , Yumi Hakozaki , Hidehiro Hojo , Takeshi Fujisawa , Riu Yamashita , Hiroshi Haeno , Akinori Kanai , Yutaka Suzuki , Masayuki Yamaguchi , Katsuya Tsuchihara , Sadamoto Zenda","doi":"10.1016/j.cllc.2025.09.013","DOIUrl":"10.1016/j.cllc.2025.09.013","url":null,"abstract":"<div><h3>Purpose</h3><div>This study aimed to evaluate whether pretreatment circulating tumor DNA (ctDNA) is a predictive biomarker for recurrence in patients with early-stage non-small cell lung cancer (ES-NSCLC), including those with pathologically unproven (PU) who received ablative radiotherapy.</div></div><div><h3>Materials and methods</h3><div>Patients clinically diagnosed with cT1-2N0M0 NSCLC treated with radiotherapy alone at a single institute were included. Pretreatment plasma and matched white blood cell samples were subjected to targeted sequencing to detect ctDNA. The association between ctDNA detection and prognosis was evaluated. Integrated analysis including non-squamous cell carcinoma (SQ) cohort and enhancement of recurrence prediction with radiographic features were also evaluated.</div></div><div><h3>Results</h3><div>Of the 19 patients (10 SQ, and 9 PU) who met the sequence quality criteria, ctDNA was detected in 6. At the median follow-up of 60 months, detectable ctDNA was significantly associated with inferior progression-free survival (PFS) in both the SQ and PU cohorts (<em>P</em> = .01 and .02). In an analysis of 62 patients combined with the non-SQ cohort, ctDNA detection was the only significant factor for PFS. The combination of ctDNA and a high positron emission tomography maximum standardized uptake value provided a better recurrence risk model than ctDNA alone (the C-index improved from 0.63 to 0.72).</div></div><div><h3>Conclusion</h3><div>These results suggest that pretreatment ctDNA detection can predict recurrence in patients with cT1-2N0M0 NSCLC, including those with PU tumors treated with ablative radiotherapy.</div></div>","PeriodicalId":10490,"journal":{"name":"Clinical lung cancer","volume":"26 8","pages":"Pages 671-679.e2"},"PeriodicalIF":3.3,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145353991","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}