Pub Date : 2024-08-11DOI: 10.1016/j.lungcan.2024.107930
Ying Li , Fangfang Xie , Qiang Zheng , Yujun Zhang , Wei Li , Minjie Xu , Qiye He , Yuan Li , Jiayuan Sun
Background
With the popularization of computed tomography, more and more pulmonary nodules (PNs) are being detected. Risk stratification of PNs is essential for detecting early-stage lung cancer while minimizing the overdiagnosis of benign nodules. This study aimed to develop a circulating tumor DNA (ctDNA) methylation-based, non-invasive model for the risk stratification of PNs.
Methods
A blood-based assay (“LUNG-TRAC”) was designed to include novel lung cancer ctDNA methylation markers identified from in-house reduced representative bisulfite sequencing data and known markers from the literature. A stratification model was trained based on 183 ctDNA samples derived from patients with benign or malignant PNs and validated in 62 patients. LUNG-TRAC was further single-blindly tested in a single- and multi-center cohort.
Results
The LUNG-TRAC model achieved an area under the curve (AUC) of 0.810 (sensitivity = 74.4 % and specificity = 73.7 %) in the validation set. Two test sets were used to evaluate the performance of LUNG-TRAC, with an AUC of 0.815 in the single-center test (N = 61; sensitivity = 67.5 % and specificity = 76.2 %) and 0.761 in the multi-center test (N = 95; sensitivity = 50.7 % and specificity = 80.8 %). The clinical utility of LUNG-TRAC was further assessed by comparing it to two established risk stratification models: the Mayo Clinic and Veteran Administration models. It outperformed both in the validation and the single-center test sets.
Conclusion
The LUNG-TRAC model demonstrated accuracy and consistency in stratifying PNs for the risk of malignancy, suggesting its utility as a non-invasive diagnostic aid for early-stage peripheral lung cancer.
{"title":"Non-invasive diagnosis of pulmonary nodules by circulating tumor DNA methylation: A prospective multicenter study","authors":"Ying Li , Fangfang Xie , Qiang Zheng , Yujun Zhang , Wei Li , Minjie Xu , Qiye He , Yuan Li , Jiayuan Sun","doi":"10.1016/j.lungcan.2024.107930","DOIUrl":"10.1016/j.lungcan.2024.107930","url":null,"abstract":"<div><h3>Background</h3><p>With the popularization of computed tomography, more and more pulmonary nodules (PNs) are being detected. Risk stratification of PNs is essential for detecting early-stage lung cancer while minimizing the overdiagnosis of benign nodules. This study aimed to develop a circulating tumor DNA (ctDNA) methylation-based, non-invasive model for the risk stratification of PNs.</p></div><div><h3>Methods</h3><p>A blood-based assay (“LUNG-TRAC”) was designed to include novel lung cancer ctDNA methylation markers identified from in-house reduced representative bisulfite sequencing data and known markers from the literature. A stratification model was trained based on 183 ctDNA samples derived from patients with benign or malignant PNs and validated in 62 patients. LUNG-TRAC was further single-blindly tested in a single- and multi-center cohort.</p></div><div><h3>Results</h3><p>The LUNG-TRAC model achieved an area under the curve (AUC) of 0.810 (sensitivity = 74.4 % and specificity = 73.7 %) in the validation set. Two test sets were used to evaluate the performance of LUNG-TRAC, with an AUC of 0.815 in the single-center test (N = 61; sensitivity = 67.5 % and specificity = 76.2 %) and 0.761 in the multi-center test (N = 95; sensitivity = 50.7 % and specificity = 80.8 %). The clinical utility of LUNG-TRAC was further assessed by comparing it to two established risk stratification models: the Mayo Clinic and Veteran Administration models. It outperformed both in the validation and the single-center test sets.</p></div><div><h3>Conclusion</h3><p>The LUNG-TRAC model demonstrated accuracy and consistency in stratifying PNs for the risk of malignancy, suggesting its utility as a non-invasive diagnostic aid for early-stage peripheral lung cancer.</p></div><div><h3>Clinical trial registration</h3><p><span><span>www.clinicaltrials.gov</span><svg><path></path></svg></span> (NCT03989219).</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107930"},"PeriodicalIF":4.5,"publicationDate":"2024-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0169500224004641/pdfft?md5=b89dd581a2a735dceff419eabb3bd3f8&pid=1-s2.0-S0169500224004641-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141984992","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-10DOI: 10.1016/j.lungcan.2024.107926
Adam Barsouk , Omar Elghawy , Alec Heidlauf , Connie Yu , Lucy Wang , David Yang , Martin Kurian , Keshav Goel , Lynn Rushkin , Anna Anran Huang , Lauren Reed-Guy , Benjamin Bleiberg , Lova Sun , Aditi Singh , Roger B. Cohen , Charu Aggarwal , Melina Marmarelis , Corey Langer
Objectives
Limited data are available comparing the efficacy of osi versus earlier generation TKIs for mNSCLC with atypical EGFR mutations (AMs) such as L861Q, G719X, S768I and exon20.
Methods
We performed a single-institution retrospective analysis of patients with EGFR-mutated mNSCLC treated from 2007 to 2023 with 1L TKIs, comparing outcomes for AM patients treated with osi, afatinib, and erlotinib. Baseline demographics, disease characteristics, treatment history, toxicity, and clinical outcomes were abstracted from the electronic medical record and compared between TKIs using independent sample t-tests and chi-square analyses. Median progression free survival (mPFS) and overall survival (mOS) were compared via Kaplan-Meier log-rank analysis and Cox multivariable regression.
Results
Among 355 patients with EGFR-mutated mNSCLC, 36 (10 %) harbored AMs in G719X (N=21; 6 %), Exon 20 (N=11; 3 %), L861Q (N=7; 2 %), S768I (N=4; 1 %), C797S (N=1; 0.3 %); 6 patients had compound mutations. Patients with classical mutations (CMs) vs AMs had similar baseline demographic and disease characteristics and usage of TKIs (p = 0.124). Among AM patients, osi yielded superior mPFS (22 m) vs afatinib (12 m; p = 0.005) or erlotinib (9 m; p = 0.001). mOS was likewise superior for osi (32 m) vs afatinib (21 m; p = 0.032) or erlotinib (17 m; p = 0.011). Dose-reduction rates due to AEs were lower for osi (19 %) vs afatinib (24 %; p = 0.003) or erlotinib (23 %; p = 0.002). Discontinuation rates due to AEs were lower for osi vs afatinib (1 % vs 2 %; p < 0.001) or erlotinib (2 %; p = 0.004).
Conclusions
In a large real-world analysis, osi demonstrated superior progression-free and overall survival and improved tolerability compared to afatinib or erlotinib for atypical EGFR-mutated mNSCLC.
{"title":"Real-world outcomes of atypical EGFR-mutated metastatic non-small cell lung cancer (mNSCLC) treated with osimertinib (osi) vs. Afatinib or erlotinib","authors":"Adam Barsouk , Omar Elghawy , Alec Heidlauf , Connie Yu , Lucy Wang , David Yang , Martin Kurian , Keshav Goel , Lynn Rushkin , Anna Anran Huang , Lauren Reed-Guy , Benjamin Bleiberg , Lova Sun , Aditi Singh , Roger B. Cohen , Charu Aggarwal , Melina Marmarelis , Corey Langer","doi":"10.1016/j.lungcan.2024.107926","DOIUrl":"10.1016/j.lungcan.2024.107926","url":null,"abstract":"<div><h3>Objectives</h3><p>Limited data are available comparing the efficacy of osi versus earlier generation TKIs for mNSCLC with atypical <em>EGFR</em> mutations (AMs) such as L861Q, G719X, S768I and exon20.</p></div><div><h3>Methods</h3><p>We performed a single-institution retrospective analysis of patients with <em>EGFR</em>-mutated mNSCLC treated from 2007 to 2023 with 1L TKIs, comparing outcomes for AM patients treated with osi, afatinib, and erlotinib. Baseline demographics, disease characteristics, treatment history, toxicity, and clinical outcomes were abstracted from the electronic medical record and compared between TKIs using independent sample t-tests and chi-square analyses. Median progression free survival (mPFS) and overall survival (mOS) were compared via Kaplan-Meier log-rank analysis and Cox multivariable regression.</p></div><div><h3>Results</h3><p>Among 355 patients with <em>EGFR</em>-mutated mNSCLC, 36 (10 %) harbored AMs in G719X (N=21; 6 %), Exon 20 (N=11; 3 %), L861Q (N=7; 2 %), S768I (N=4; 1 %), C797S (N=1; 0.3 %); 6 patients had compound mutations. Patients with classical mutations (CMs) vs AMs had similar baseline demographic and disease characteristics and usage of TKIs (p = 0.124). Among AM patients, osi yielded superior mPFS (22 m) vs afatinib (12 m; p = 0.005) or erlotinib (9 m; p = 0.001). mOS was likewise superior for osi (32 m) vs afatinib (21 m; p = 0.032) or erlotinib (17 m; p = 0.011). Dose-reduction rates due to AEs were lower for osi (19 %) vs afatinib (24 %; p = 0.003) or erlotinib (23 %; p = 0.002). Discontinuation rates due to AEs were lower for osi vs afatinib (1 % vs 2 %; p < 0.001) or erlotinib (2 %; p = 0.004).</p></div><div><h3>Conclusions</h3><p>In a large real-world analysis, osi demonstrated superior progression-free and overall survival and improved tolerability compared to afatinib or erlotinib for atypical <em>EGFR</em>-mutated mNSCLC.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107926"},"PeriodicalIF":4.5,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141954230","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-10DOI: 10.1016/j.lungcan.2024.107924
Martin Reck , Rafal Dziadziuszko , Shunichi Sugawara , Steven Kao , Maximilian Hochmair , Florian Huemer , Gilberto de Castro Jr , Libor Havel , Reyes Bernabé Caro , György Losonczy , Jong-Seok Lee , Dariusz M. Kowalski , Zoran Andric , Raffaele Califano , Andrea Veatch , Gregory Gerstner , Marta Batus , Stefanie Morris , Monika Kaul , Vaikunth Cuchelkar , Stephen V. Liu
Objectives
IMbrella A is a Phase III extension study that allowed rollover from Roche/Genentech–sponsored atezolizumab trials, including IMpower133, a Phase I/III trial of first-line atezolizumab or placebo plus carboplatin/etoposide in extensive-stage small cell lung cancer. We report outcomes from an exploratory analysis of IMpower133 with extended time-to-event data for patients who rolled over to IMbrella A.
Materials and Methods
IMpower133 patients could roll over to IMbrella A to receive atezolizumab 1200 mg intravenously every three weeks if they continued to receive atezolizumab at IMpower133 closure or were in survival follow-up after atezolizumab discontinuation. Overall survival and safety were assessed; only serious adverse events and AEs of special interest were collected in IMbrella A.
Results
Eighteen of 26 eligible patients rolled over to IMbrella A. At clinical cutoff (March 16, 2023), median follow-up in the atezolizumab plus carboplatin/etoposide arm (IMpower133 and IMbrella A) was 59.4 months. The three-, four-, and five-year overall survival (95 % CI) estimates were 16 % (11 %–21 %), 13 % (8 %–18 %), and 12 % (7 %–17 %), respectively. In IMbrella A, serious adverse events occurred in three patients (16.7 %), and one adverse event of special interest was reported (grade two hypothyroidism).
Conclusion
This long-term analysis of patients from IMbrella A previously enrolled in IMpower133 provides the first report of five-year overall survival outcomes in patients with extensive-stage small cell lung cancer treated with first-line cancer immunotherapy and chemotherapy. While limited by small patient numbers and lack of long-term data for the IMpower133 control arm, exploratory overall survival analyses in patients treated with atezolizumab plus carboplatin/etoposide compared favorably with historical data with chemotherapy alone.
{"title":"Five-year survival in patients with extensive-stage small cell lung cancer treated with atezolizumab in the Phase III IMpower133 study and the Phase III IMbrella A extension study","authors":"Martin Reck , Rafal Dziadziuszko , Shunichi Sugawara , Steven Kao , Maximilian Hochmair , Florian Huemer , Gilberto de Castro Jr , Libor Havel , Reyes Bernabé Caro , György Losonczy , Jong-Seok Lee , Dariusz M. Kowalski , Zoran Andric , Raffaele Califano , Andrea Veatch , Gregory Gerstner , Marta Batus , Stefanie Morris , Monika Kaul , Vaikunth Cuchelkar , Stephen V. Liu","doi":"10.1016/j.lungcan.2024.107924","DOIUrl":"10.1016/j.lungcan.2024.107924","url":null,"abstract":"<div><h3>Objectives</h3><p>IMbrella A is a Phase III extension study that allowed rollover from Roche/Genentech–sponsored atezolizumab trials, including IMpower133, a Phase I/III trial of first-line atezolizumab or placebo plus carboplatin/etoposide in extensive-stage small cell lung cancer. We report outcomes from an exploratory analysis of IMpower133 with extended time-to-event data for patients who rolled over to IMbrella A.</p></div><div><h3>Materials and Methods</h3><p>IMpower133 patients could roll over to IMbrella A to receive atezolizumab 1200 mg intravenously every three weeks if they continued to receive atezolizumab at IMpower133 closure or were in survival follow-up after atezolizumab discontinuation. Overall survival and safety were assessed; only serious adverse events and AEs of special interest were collected in IMbrella A.</p></div><div><h3>Results</h3><p>Eighteen of 26 eligible patients rolled over to IMbrella A. At clinical cutoff (March 16, 2023), median follow-up in the atezolizumab plus carboplatin/etoposide arm (IMpower133 and IMbrella A) was 59.4 months. The three-, four-, and five-year overall survival (95 % CI) estimates were 16 % (11 %–21 %), 13 % (8 %–18 %), and 12 % (7 %–17 %), respectively. In IMbrella A, serious adverse events occurred in three patients (16.7 %), and one adverse event of special interest was reported (grade two hypothyroidism).</p></div><div><h3>Conclusion</h3><p>This long-term analysis of patients from IMbrella A previously enrolled in IMpower133 provides the first report of five-year overall survival outcomes in patients with extensive-stage small cell lung cancer treated with first-line cancer immunotherapy and chemotherapy. While limited by small patient numbers and lack of long-term data for the IMpower133 control arm, exploratory overall survival analyses in patients treated with atezolizumab plus carboplatin/etoposide compared favorably with historical data with chemotherapy alone.</p><p>NCT03148418.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"196 ","pages":"Article 107924"},"PeriodicalIF":4.5,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142274412","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-10DOI: 10.1016/j.lungcan.2024.107927
Julia Naso , Aakash Desai , Caleb J. Smith , Yash P. Ashara , Stephen Yip , Ying-Chun Lo
Objectives
Accurately predicting which patients diagnosed with non-small cell lung cancer (NSCLC) will respond to immunotherapy remains a clinical challenge. This study aims to determine the associations between MYC immunoreactivity, MYC copy number gain (CNG), driver mutations and survival following immunotherapy treatment, to provide insight into whether clinical MYC assessment may have predictive value.
Materials and Methods
MYC copy number status was determined in 82 patients with NSCLC treated with immunotherapy, and MYC immunohistochemistry (IHC) was performed on 80 of these cases. MYC staining in ≥ 40 % of tumor cells was considered positive. Driver gene alterations, PD-L1 status and survival outcomes were assessed through retrospective chart review. Overall survival (OS) and progression free survival (PFS) were calculated from the date of immunotherapy initiation.
Results
Nine (11 %) of 82 cases had MYC CNG and 56 (70 %) of the 80 immunostained cases were positive for MYC. MYC CNG was significantly associated with STK11 mutation (P=0.023), whereas positive MYC IHC was significantly associated with KRAS mutation (P=0.0076) and current/former smoking (P=0.0007). MYC CNG and positive MYC IHC were not significantly associated with each other (P=0.42), or with PD-L1 ≥ 1 % (MYC CNG: P=0.10; MYC IHC: P=0.09). Positive MYC IHC and PD-L1 ≥ 1 % were both significant predictors of OS (MYC: HR 2.7, 95 % CI 1.1–6.4, P=0.026; PD-L1: HR 0.33, 95 % CI 0.15–0.72, P=0.0055). MYC IHC positive/PD-L1 < 1 % cases had the shortest OS (median 230 versus 918 days, P=0.00069) and PFS (median 84 versus 254 days, P=0.0087). MYC CNG was not associated with OS or PFS.
Conclusion
We find that positive MYC IHC is an independent predictor of shorter OS after immunotherapy treatment, with MYC positive/PD-L1 < 1 % status predictive of particularly poor immunotherapy response. We identify positive MYC IHC as a feature of possible relevance to NSCLC treatment selection and of interest for future therapy development.
{"title":"Predictive value and molecular correlates of MYC immunohistochemistry and copy number gain in non-small cell lung carcinomas treated with immunotherapy","authors":"Julia Naso , Aakash Desai , Caleb J. Smith , Yash P. Ashara , Stephen Yip , Ying-Chun Lo","doi":"10.1016/j.lungcan.2024.107927","DOIUrl":"10.1016/j.lungcan.2024.107927","url":null,"abstract":"<div><h3>Objectives</h3><p>Accurately predicting which patients diagnosed with non-small cell lung cancer (NSCLC) will respond to immunotherapy remains a clinical challenge. This study aims to determine the associations between MYC immunoreactivity, <em>MYC</em> copy number gain (CNG), driver mutations and survival following immunotherapy treatment, to provide insight into whether clinical MYC assessment may have predictive value.</p></div><div><h3>Materials and Methods</h3><p>MYC copy number status was determined in 82 patients with NSCLC treated with immunotherapy, and MYC immunohistochemistry (IHC) was performed on 80 of these cases. MYC staining in ≥ 40 % of tumor cells was considered positive. Driver gene alterations, PD-L1 status and survival outcomes were assessed through retrospective chart review. Overall survival (OS) and progression free survival (PFS) were calculated from the date of immunotherapy initiation.</p></div><div><h3>Results</h3><p>Nine (11 %) of 82 cases had <em>MYC</em> CNG and 56 (70 %) of the 80 immunostained cases were positive for MYC. <em>MYC</em> CNG was significantly associated with <em>STK11</em> mutation (P=0.023), whereas positive MYC IHC was significantly associated with <em>KRAS</em> mutation (P=0.0076) and current/former smoking (P=0.0007). <em>MYC</em> CNG and positive MYC IHC were not significantly associated with each other (P=0.42), or with PD-L1 ≥ 1 % (MYC CNG: P=0.10; MYC IHC: P=0.09). Positive MYC IHC and PD-L1 ≥ 1 % were both significant predictors of OS (MYC: HR 2.7, 95 % CI 1.1–6.4, P=0.026; PD-L1: HR 0.33, 95 % CI 0.15–0.72, P=0.0055). MYC IHC positive/PD-L1 < 1 % cases had the shortest OS (median 230 versus 918 days, P=0.00069) and PFS (median 84 versus 254 days, P=0.0087). <em>MYC</em> CNG was not associated with OS or PFS.</p></div><div><h3>Conclusion</h3><p>We find that positive MYC IHC is an independent predictor of shorter OS after immunotherapy treatment, with MYC positive/PD-L1 < 1 % status predictive of particularly poor immunotherapy response. We identify positive MYC IHC as a feature of possible relevance to NSCLC treatment selection and of interest for future therapy development.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107927"},"PeriodicalIF":4.5,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142011518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-09DOI: 10.1016/j.lungcan.2024.107920
Virginia Calvo , Emetis Niazmand , Enric Carcereny , Delvys Rodriguez-Abreu , Manuel Cobo , Rafael López-Castro , María Guirado , Carlos Camps , Ana Laura Ortega , Reyes Bernabé , Bartomeu Massutí , Rosario Garcia-Campelo , Edel del Barco , José Luis González-Larriba , Joaquim Bosch-Barrera , Marta Martínez , María Torrente , María-Esther Vidal , Mariano Provencio
Objectives
Lung Cancer (LC) is a multifactorial disease for which the role of genetic susceptibility has become increasingly relevant. Our aim was to use artificial intelligence (AI) to analyze differences between patients with LC based on family history of cancer (FHC).
Materials and methods
From August 2016 to June 2020 clinical information was obtained from Thoracic Tumors Registry (TTR), a nationwide database sponsored by the Spanish Lung Cancer Group. In addition to descriptive statistical analysis, an AI-assisted analysis was performed. The German Technical Information Library supported the merging of data from the electronic medical records and database of the TTR. The results of the AI-assisted analysis were reported using Knowledge Graph, Unified Schema and descriptive and predictive analyses.
Results
Analyses were performed in two phases: first, conventional statistical analysis including 11,684 patients of those 5,806 had FHC. Median overall survival (OS) for the global population was 23 months (CI 95 %: 21.39–24.61) in patients with FHC versus 21 months (CI 95 %: 19.53–22.48) in patients without FHC (NFHC), p < 0.001. The second AI-assisted analysis included 5,788 patients of those 939 had FHC. 58.48 % of women with FHC had LC. 9.53 % of patients had an EGFR or HER2 mutation or ALK translocation and at least one relative with cancer. A family history of LC was associated with an increased risk of smoking-related LC. Non-smokers with a family history of LC were more likely to have an EGFR mutation in NSCLC. In Bayesian network analysis, 55 % of patients with a family history of LC and never-smokers had an EGFR mutation.
Conclusion
In our population, the incidence of LC in patients with a FHC is higher in women and younger patients. FHC is a risk factor and predictor of LC development, especially in people ≤ 50 years. These results were confirmed by conventional statistics and AI-assisted analysis.
{"title":"Family history of cancer and lung cancer: Utility of big data and artificial intelligence for exploring the role of genetic risk","authors":"Virginia Calvo , Emetis Niazmand , Enric Carcereny , Delvys Rodriguez-Abreu , Manuel Cobo , Rafael López-Castro , María Guirado , Carlos Camps , Ana Laura Ortega , Reyes Bernabé , Bartomeu Massutí , Rosario Garcia-Campelo , Edel del Barco , José Luis González-Larriba , Joaquim Bosch-Barrera , Marta Martínez , María Torrente , María-Esther Vidal , Mariano Provencio","doi":"10.1016/j.lungcan.2024.107920","DOIUrl":"10.1016/j.lungcan.2024.107920","url":null,"abstract":"<div><h3>Objectives</h3><p>Lung Cancer (LC) is a multifactorial disease for which the role of genetic susceptibility has become increasingly relevant. Our aim was to use artificial intelligence (AI) to analyze differences between patients with LC based on family history of cancer (FHC).</p></div><div><h3>Materials and methods</h3><p>From August 2016 to June 2020 clinical information was obtained from Thoracic Tumors Registry (TTR), a nationwide database sponsored by the Spanish Lung Cancer Group. In addition to descriptive statistical analysis, an AI-assisted analysis was performed. The German Technical Information Library supported the merging of data from the electronic medical records and database of the TTR. The results of the AI-assisted analysis were reported using Knowledge Graph, Unified Schema and descriptive and predictive analyses.</p></div><div><h3>Results</h3><p>Analyses were performed in two phases: first, conventional statistical analysis including 11,684 patients of those 5,806 had FHC. Median overall survival (OS) for the global population was 23 months (CI 95 %: 21.39–24.61) in patients with FHC versus 21 months (CI 95 %: 19.53–22.48) in patients without FHC (NFHC), p < 0.001. The second AI-assisted analysis included 5,788 patients of those 939 had FHC. 58.48 % of women with FHC had LC. 9.53 % of patients had an EGFR or HER2 mutation or ALK translocation and at least one relative with cancer. A family history of LC was associated with an increased risk of smoking-related LC. Non-smokers with a family history of LC were more likely to have an EGFR mutation in NSCLC. In Bayesian network analysis, 55 % of patients with a family history of LC and never-smokers had an EGFR mutation.</p></div><div><h3>Conclusion</h3><p>In our population, the incidence of LC in patients with a FHC is higher in women and younger patients. FHC is a risk factor and predictor of LC development, especially in people ≤ 50 years. These results were confirmed by conventional statistics and AI-assisted analysis.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107920"},"PeriodicalIF":4.5,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0169500224004549/pdfft?md5=97d1b85b57d7a4c02403157543ffb63f&pid=1-s2.0-S0169500224004549-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141976099","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-08DOI: 10.1016/j.lungcan.2024.107925
Yao Ding , Shun Lei , Ling Wang , Long Tang , Yue Zhang , Yiran Liao , Xia Deng , Yan Li , Yi Gong , Yongsheng Li
Objective
The reported impact of age on the effectiveness of emerging immunotherapies in patients with advanced non-small cell lung cancer (NSCLC) has been inconsistent in clinical trials, largely due to an underrepresentation of older individuals. This meta-analysis aimed to evaluate the efficacy of immune checkpoint inhibitor (ICI) in older patients with NSCLC.
Materials and methods
The literature up to April 2024 was reviewed to identify articles meeting the criteria for inclusion. Hazard ratios (HRs) for overall survival (OS) across various age groups were examined. The ratio of HR (RHR) was computed and combined for each study.
Results
A preliminary search identified 118 articles, with 13 being phase II or III randomized clinical trials comparing the efficacy of nivolumab, avelumab, ipilimumab, pembrolizumab, atezolizumab, and chemotherapy with or without antiangiogenic therapy. The analysis revealed that the HR for OS was 0.75 (95 % CI: 0.70–0.80, P=0.080) in patients aged under 75 years and 0.87 (95 % CI: 0.74–1.01, P=0.913) in patients aged 75 years and older. The combined RHR for patients aged 75 years and above versus those aged under 75 years was 1.14 (95 % CI: 0.97–1.34, P=0.697). There was no significant difference in OS benefit between patients over 75 years and younger patients (P=0.105). Subgroup analyses indicated that the benefit of OS was consistent across all subgroups and age groups.
Conclusions
Our investigation found no significant differences in the efficacy of immunotherapy for patients with NSCLC aged 75 years and older compared to those under 75 years old. This suggests that the efficacy of immunotherapy against NSCLC is consistent across age groups.
{"title":"Age-related efficacy of immunotherapies in advanced non-small cell lung cancer: a comprehensive meta-analysis","authors":"Yao Ding , Shun Lei , Ling Wang , Long Tang , Yue Zhang , Yiran Liao , Xia Deng , Yan Li , Yi Gong , Yongsheng Li","doi":"10.1016/j.lungcan.2024.107925","DOIUrl":"10.1016/j.lungcan.2024.107925","url":null,"abstract":"<div><h3>Objective</h3><p>The reported impact of age on the effectiveness of emerging immunotherapies in patients with advanced non-small cell lung cancer (NSCLC) has been inconsistent in clinical trials, largely due to an underrepresentation of older individuals. This <em>meta</em>-analysis aimed to evaluate the efficacy of immune checkpoint inhibitor (ICI) in older patients with NSCLC.</p></div><div><h3>Materials and methods</h3><p>The literature up to April 2024 was reviewed to identify articles meeting the criteria for inclusion. Hazard ratios (HRs) for overall survival (OS) across various age groups were examined. The ratio of HR (RHR) was computed and combined for each study.</p></div><div><h3>Results</h3><p>A preliminary search identified 118 articles, with 13 being phase II or III randomized clinical trials comparing the efficacy of nivolumab, avelumab, ipilimumab, pembrolizumab, atezolizumab, and chemotherapy with or without antiangiogenic therapy. The analysis revealed that the HR for OS was 0.75 (95 % CI: 0.70–0.80, <em>P</em>=0.080) in patients aged under 75 years and 0.87 (95 % CI: 0.74–1.01, <em>P</em>=0.913) in patients aged 75 years and older. The combined RHR for patients aged 75 years and above <em>versus</em> those aged under 75 years was 1.14 (95 % CI: 0.97–1.34, <em>P</em>=0.697). There was no significant difference in OS benefit between patients over 75 years and younger patients (<em>P</em>=0.105). Subgroup analyses indicated that the benefit of OS was consistent across all subgroups and age groups.</p></div><div><h3>Conclusions</h3><p>Our investigation found no significant differences in the efficacy of immunotherapy for patients with NSCLC aged 75 years and older compared to those under 75 years old. This suggests that the efficacy of immunotherapy against NSCLC is consistent across age groups.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107925"},"PeriodicalIF":4.5,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141988280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-06DOI: 10.1016/j.lungcan.2024.107921
David M. Waterhouse , Sacha Rothschild , Christophe Dooms , Bertrand Mennecier , Farastuk Bozorgmehr , Margarita Majem , Michel H. van den Heuvel , Helena Linardou , Byoung Chul Cho , Rachel Roberts-Thomson , Kentaro Tanaka , Normand Blais , Gustavo Schvartsman , Karin Holmskov Hansen , Izabela Chmielewska , Martin D. Forster , Christina Giannopoulou , Björn Stollenwerk , Cynthia C. Obiozor , Yang Wang , Silvia Novello
Background
In the CodeBreaK 200 phase III, open-label trial, sotorasib significantly improved efficacy versus docetaxel in previously treated KRAS G12C-mutated advanced non-small cell lung cancer (NSCLC). Patient-reported outcomes (PROs) for global health status, physical functioning, dyspnea, and cough favored sotorasib over docetaxel. Here, we report sotorasib’s additional impact on quality of life (QOL).
Methods
In CodeBreaK 200, 345 patients who had progressed after prior therapy received sotorasib (960 mg orally daily) or docetaxel (75 mg/m2 intravenously every 3 weeks). Validated questionnaires captured patients’ perception of their QOL and symptom burden for key secondary and exploratory PRO endpoints, including the European Organisation for Research and Treatment of Cancer Quality-of-life Questionnaire Core 30 (EORTC QLQ-C30) and Quality-of-life Questionnaire Lung Cancer 13 (EORTC QLQ-LC13), question GP5 from the Functional Assessment of Cancer Therapy Tool General Form (FACT-G GP5), PRO-Common Terminology Criteria for Adverse Events (PRO-CTCAE), and 5-level EuroQOL-5 dimensions (EQ-5D-5L) including visual analog scale (EQ-5D VAS). Change from baseline to week 12 was assessed with generalized estimating equations for ordinal outcomes.
Results
Patients receiving sotorasib were less bothered by treatment side effects than those receiving docetaxel (odds ratio [OR] 5.7) and experienced symptoms at lower severity (pain: OR 2.9; aching muscles: OR 4.4; aching joints: OR 4.2; mouth or throat sores: OR 4.3). Further, patients’ symptoms interfered less with usual/daily activities (pain: OR 3.2; aching muscles: OR 3.9; aching joints: OR 10.7). QOL remained stable with sotorasib but worsened with docetaxel (change from baseline in EQ-5D VAS score: 1.5 vs –8.4 at cycle 1 day 5 and 2.2 vs –5.8 at week 12).
Conclusions
Patients receiving sotorasib reported less severe symptoms than those receiving docetaxel. In addition to improving clinical efficacy outcomes, sotorasib maintained QOL versus docetaxel, suggesting sotorasib may be a more tolerable treatment option for patients with pretreated, KRAS G12C-mutated advanced NSCLC.
{"title":"Patient-reported outcomes in CodeBreaK 200: Sotorasib versus docetaxel for previously treated advanced NSCLC with KRAS G12C mutation","authors":"David M. Waterhouse , Sacha Rothschild , Christophe Dooms , Bertrand Mennecier , Farastuk Bozorgmehr , Margarita Majem , Michel H. van den Heuvel , Helena Linardou , Byoung Chul Cho , Rachel Roberts-Thomson , Kentaro Tanaka , Normand Blais , Gustavo Schvartsman , Karin Holmskov Hansen , Izabela Chmielewska , Martin D. Forster , Christina Giannopoulou , Björn Stollenwerk , Cynthia C. Obiozor , Yang Wang , Silvia Novello","doi":"10.1016/j.lungcan.2024.107921","DOIUrl":"10.1016/j.lungcan.2024.107921","url":null,"abstract":"<div><h3>Background</h3><p>In the CodeBreaK 200 phase III, open-label trial, sotorasib significantly improved efficacy versus docetaxel in previously treated <em>KRAS</em> G12C-mutated advanced non-small cell lung cancer (NSCLC). Patient-reported outcomes (PROs) for global health status, physical functioning, dyspnea, and cough favored sotorasib over docetaxel. Here, we report sotorasib’s additional impact on quality of life (QOL).</p></div><div><h3>Methods</h3><p>In CodeBreaK 200, 345 patients who had progressed after prior therapy received sotorasib (960 mg orally daily) or docetaxel (75 mg/m<sup>2</sup> intravenously every 3 weeks). Validated questionnaires captured patients’ perception of their QOL and symptom burden for key secondary and exploratory PRO endpoints, including the European Organisation for Research and Treatment of Cancer Quality-of-life Questionnaire Core 30 (EORTC QLQ-C30) and Quality-of-life Questionnaire Lung Cancer 13 (EORTC QLQ-LC13), question GP5 from the Functional Assessment of Cancer Therapy Tool General Form (FACT-G GP5), PRO-Common Terminology Criteria for Adverse Events (PRO-CTCAE), and 5-level EuroQOL-5 dimensions (EQ-5D-5L) including visual analog scale (EQ-5D VAS). Change from baseline to week 12 was assessed with generalized estimating equations for ordinal outcomes.</p></div><div><h3>Results</h3><p>Patients receiving sotorasib were less bothered by treatment side effects than those receiving docetaxel (odds ratio [OR] 5.7) and experienced symptoms at lower severity (pain: OR 2.9; aching muscles: OR 4.4; aching joints: OR 4.2; mouth or throat sores: OR 4.3). Further, patients’ symptoms interfered less with usual/daily activities (pain: OR 3.2; aching muscles: OR 3.9; aching joints: OR 10.7). QOL remained stable with sotorasib but worsened with docetaxel (change from baseline in EQ-5D VAS score: 1.5 vs –8.4 at cycle 1 day 5 and 2.2 vs –5.8 at week 12).</p></div><div><h3>Conclusions</h3><p>Patients receiving sotorasib reported less severe symptoms than those receiving docetaxel. In addition to improving clinical efficacy outcomes, sotorasib maintained QOL versus docetaxel, suggesting sotorasib may be a more tolerable treatment option for patients with pretreated, <em>KRAS</em> G12C-mutated advanced NSCLC.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"196 ","pages":"Article 107921"},"PeriodicalIF":4.5,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0169500224004550/pdfft?md5=f788b6bd78bf93b5b72d2444d5c9ed96&pid=1-s2.0-S0169500224004550-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142274408","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-05DOI: 10.1016/j.lungcan.2024.107918
Yiqun Wei , Runmiao Wu , Shuanying Yang , Yanfei Cao , Jing Li , Huihui Ma , Junfang Wu , Jinjin Duan , Shumei Yang
Objectives
Tigger transposable element-derived 1 (TIGD1) expression and its underlying functions and regulatory mechanisms in lung adenocarcinoma (LUAD) remain unknown. Therefore, we intended to explore the expression, potential functions, and regulatory mechanisms of TIGD1 in LUAD.
Materials and methods
TIGD1 expression in LUAD tissues was determined by immunohistochemistry analysis of a tissue microarray. Functional experiments were conducted to determine how TIGD1 affects LUAD tumorigenesis and metastasis. The molecular mechanisms by which TIGD1 induces LUAD progression were determined.
Results
TIGD1 was upregulated in LUAD tissues and was related to lymph node metastases. TIGD1 knockdown suppressed LUAD cell proliferation, migration, and invasion, while promoted cell apoptosis. Furthermore, decreased metastatic nodules were observed in the TIGD1 knockdown mouse metastasis model. Moreover, microarray analysis was performed to determine the potential downstream genes of TIGD1 in LUAD. Hallmark pathway analysis revealed that the downstream genes of TIGD1 were involved in epithelial-mesenchymal transition (EMT). Western blotting confirmed that vimentin and TWIST was downregulated in TIGD1 knockdown cells, while E-cadherin was upregulated. Ingenuity pathway and hallmark pathway analyses revealed that TIGD1 regulated the interleukin-6 signaling pathway and related gene members. Western blotting, quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay indicated that downregulation of TIGD1 decreased interleukin-6 and CXCL1 expression. TIGD1 expression was negatively correlated with immune infiltration in LUAD. The upstream microRNA of TIGD1 was predicted, and subsequent luciferase reporter gene experiments confirmed the interactions between miR-137 and TIGD1. The expression of miR-137 was significantly downregulated in LUAD tissues and miR-137 suppressed the proliferation, migration, and invasion of LUAD cells, partially through negatively regulating the expression of TIGD1.
Conclusion
Our findings suggest that TIGD1, which was regulated by miR-137, contributed to LUAD progression by promoting cell proliferation, migration, invasion, and EMT and suppressing cell apoptosis.
{"title":"MiR-137 mediated high expression of TIGD1 promotes migration, invasion, and suppresses apoptosis of lung adenocarcinoma","authors":"Yiqun Wei , Runmiao Wu , Shuanying Yang , Yanfei Cao , Jing Li , Huihui Ma , Junfang Wu , Jinjin Duan , Shumei Yang","doi":"10.1016/j.lungcan.2024.107918","DOIUrl":"10.1016/j.lungcan.2024.107918","url":null,"abstract":"<div><h3>Objectives</h3><p>Tigger transposable element-derived 1 (TIGD1) expression and its underlying functions and regulatory mechanisms in lung adenocarcinoma (LUAD) remain unknown. Therefore, we intended to explore the expression, potential functions, and regulatory mechanisms of TIGD1 in LUAD.</p></div><div><h3>Materials and methods</h3><p>TIGD1 expression in LUAD tissues was determined by immunohistochemistry analysis of a tissue microarray. Functional experiments were conducted to determine how TIGD1 affects LUAD tumorigenesis and metastasis. The molecular mechanisms by which TIGD1 induces LUAD progression were determined.</p></div><div><h3>Results</h3><p>TIGD1 was upregulated in LUAD tissues and was related to lymph node metastases. TIGD1 knockdown suppressed LUAD cell proliferation, migration, and invasion, while promoted cell apoptosis. Furthermore, decreased metastatic nodules were observed in the TIGD1 knockdown mouse metastasis model. Moreover, microarray analysis was performed to determine the potential downstream genes of TIGD1 in LUAD. Hallmark pathway analysis revealed that the downstream genes of TIGD1 were involved in epithelial-mesenchymal transition (EMT). Western blotting confirmed that vimentin and TWIST was downregulated in TIGD1 knockdown cells, while E-cadherin was upregulated. Ingenuity pathway and hallmark pathway analyses revealed that TIGD1 regulated the interleukin-6 signaling pathway and related gene members. Western blotting, quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay indicated that downregulation of TIGD1 decreased interleukin-6 and CXCL1 expression. TIGD1 expression was negatively correlated with immune infiltration in LUAD. The upstream microRNA of TIGD1 was predicted, and subsequent luciferase reporter gene experiments confirmed the interactions between miR-137 and TIGD1. The expression of miR-137 was significantly downregulated in LUAD tissues and miR-137 suppressed the proliferation, migration, and invasion of LUAD cells, partially through negatively regulating the expression of TIGD1.</p></div><div><h3>Conclusion</h3><p>Our findings suggest that TIGD1, which was regulated by miR-137, contributed to LUAD progression by promoting cell proliferation, migration, invasion, and EMT and suppressing cell apoptosis.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107918"},"PeriodicalIF":4.5,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0169500224004525/pdfft?md5=1e665bf0e5a69be50ea5a3d4465969fc&pid=1-s2.0-S0169500224004525-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142020622","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-05DOI: 10.1016/j.lungcan.2024.107916
Najmeh Alirezaie , Anne-Laure Chong , Felix K.F. Kommoss , Nelly Sabbaghian , Jose Camacho Valenzuela , Dylan Pelletier , Javad Nadaf , Shailesh B. Kolekar , Pradeesh Sivapalan , Mark G. Evans , Paul S. Thorner , Pierre-Olivier Fiset , Andreas von Deimling , William D. Foulkes
Objective
Pulmonary blastoma is a rare, biphasic, adult-onset lung tumor. In this study, we investigate whether DICER1 pathogenic variants are a feature of pulmonary blastomas through in-depth analysis of the molecular events defining them.
Methods
We performed exome-wide sequencing and DNA methylation profiling of 8 pulmonary blastomas from 6 affected persons.
Results
We identified biallelic somatic DICER1 pathogenic variants in 7 of 8 cases. The remaining case had a solitary missense pathogenic variant in the RNase IIIb domain of DICER1. Six of 8 cases carried a CTNNB1 hotspot variant and 4 of 8 had a somatic pathogenic variant in TP53. Methylation analysis showed that the pulmonary blastomas clustered with other DICER1-mutated tumors and not with other more common types of lung cancer.
Conclusion
We conclude somatic DICER1 pathogenic variants are the major driver of pulmonary blastoma and are likely to act in conjunction with CTNNB1 hotspot variants that are often present.
{"title":"Exomic and epigenomic analysis of pulmonary blastoma","authors":"Najmeh Alirezaie , Anne-Laure Chong , Felix K.F. Kommoss , Nelly Sabbaghian , Jose Camacho Valenzuela , Dylan Pelletier , Javad Nadaf , Shailesh B. Kolekar , Pradeesh Sivapalan , Mark G. Evans , Paul S. Thorner , Pierre-Olivier Fiset , Andreas von Deimling , William D. Foulkes","doi":"10.1016/j.lungcan.2024.107916","DOIUrl":"10.1016/j.lungcan.2024.107916","url":null,"abstract":"<div><h3>Objective</h3><p>Pulmonary blastoma is a rare, biphasic, adult-onset lung tumor. In this study, we investigate whether DICER1 pathogenic variants are a feature of pulmonary blastomas through in-depth analysis of the molecular events defining them.</p></div><div><h3>Methods</h3><p>We performed exome-wide sequencing and DNA methylation profiling of 8 pulmonary blastomas from 6 affected persons.</p></div><div><h3>Results</h3><p>We identified biallelic somatic <em>DICER1</em> pathogenic variants in 7 of 8 cases. The remaining case had a solitary missense pathogenic variant in the RNase IIIb domain of DICER1. Six of 8 cases carried a <em>CTNNB1</em> hotspot variant and 4 of 8 had a somatic pathogenic variant in <em>TP53</em>. Methylation analysis showed that the pulmonary blastomas clustered with other <em>DICER1</em>-mutated tumors and not with other more common types of lung cancer.</p></div><div><h3>Conclusion</h3><p>We conclude somatic <em>DICER1</em> pathogenic variants are the major driver of pulmonary blastoma and are likely to act in conjunction with <em>CTNNB1</em> hotspot variants that are often present.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107916"},"PeriodicalIF":4.5,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141913176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Several patients treated with osimertinib experience progressive disease. The aim was to clarify the mechanisms underlying resistance to osimertinib.
Methods
ELUCIDATOR: A multi-centre, prospective, observational study involved chemotherapy-naive patients with advanced non-small cell lung cancer receiving osimertinib. Mutations in cancer-associated genes, detected via ultrasensitive next-generation sequencing of circulating tumour deoxyribonucleic acid samples, were collected at baseline and after progressive disease detection. These paired plasma samples were compared.
Results
Of 188 patients enrolled (May 2019-January 2021), 178 (119 females [67 %]) median age 74 years, were included. Patients, n = 95 (53 %) had epidermal growth factor receptor exon 19 deletion mutations. Among 115 patients with progressive disease, circulating tumour deoxyribonucleic acid levels of 85 patients were analysed. MET amplification (n = 4), TP53 mutations (n = 4), PIK3CA mutations (n = 3), BRINP3 mutation (n = 2), BRAF mutation (n = 2), APC mutation (n = 1), RET mutation (n = 1) and epidermal growth factor receptor (EGFR) resistance mutation, and C797S (n = 1) were detected. Patients with baseline TP53 mutations, with MET or EGFR amplification had shorter progression-free (PFS) and overall survival. Patients with PIK3CA mutations tended to shorter PFS.
Conclusion
MET amplification and PIK3CA mutation mechanisms underly resistance to osimertinib in patients. Patients with coexisting mutations or amplifications at baseline had shorter PFS and overall survival.
{"title":"Mechanisms of resistance and correlation between pre-treatment co-alterations and p-prognosis to osimertinib in chemo-naïve advanced non-small cell lung cancer","authors":"Akihiro Tamiya , Mitsuo Osuga , Daijiro Harada , Shun-ichi Isa , Yoshihiko Taniguchi , Keiichi Nakamura , Yasuyuki Mizumori , Tsutomu Shinohara , Hidetoshi Yanai , Katsumi Nakatomi , Masahide Oki , Masahide Mori , Tomohito Kuwako , Koji Yamazaki , Atsuhisa Tamura , Masahiko Ando , Yasuhiro Koh","doi":"10.1016/j.lungcan.2024.107917","DOIUrl":"10.1016/j.lungcan.2024.107917","url":null,"abstract":"<div><h3>Background</h3><p>Several patients treated with osimertinib experience progressive disease. The aim was to clarify the mechanisms underlying resistance to osimertinib.</p></div><div><h3>Methods</h3><p>ELUCIDATOR: A multi-centre, prospective, observational study involved chemotherapy-naive patients with advanced non-small cell lung cancer receiving osimertinib. Mutations in cancer-associated genes, detected via ultrasensitive next-generation sequencing of circulating tumour deoxyribonucleic acid samples, were collected at baseline and after progressive disease detection. These paired plasma samples were compared.</p></div><div><h3>Results</h3><p>Of 188 patients enrolled (May 2019-January 2021), 178 (119 females [67 %]) median age 74 years, were included. Patients, n = 95 (53 %) had epidermal growth factor receptor exon 19 deletion mutations. Among 115 patients with progressive disease, circulating tumour deoxyribonucleic acid levels of 85 patients were analysed. <em>MET</em> amplification (n = 4), <em>TP53</em> mutations (n = 4), <em>PIK3CA</em> mutations (n = 3), <em>BRINP3</em> mutation (n = 2), <em>BRAF</em> mutation (n = 2), <em>APC</em> mutation (n = 1), <em>RET</em> mutation (n = 1) and epidermal growth factor receptor (EGFR) resistance mutation, and <em>C797S</em> (n = 1) were detected. Patients with baseline <em>TP53</em> mutations, with <em>MET</em> or <em>EGFR</em> amplification had shorter progression-free (PFS) and overall survival. Patients with <em>PIK3CA</em> mutations tended to shorter PFS.</p></div><div><h3>Conclusion</h3><p><em>MET</em> amplification and <em>PIK3CA</em> mutation mechanisms underly resistance to osimertinib in patients. Patients with coexisting mutations or amplifications at baseline had shorter PFS and overall survival.</p></div>","PeriodicalId":18129,"journal":{"name":"Lung Cancer","volume":"195 ","pages":"Article 107917"},"PeriodicalIF":4.5,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0169500224004513/pdfft?md5=e2dc34b44774c91db765513395ec6f32&pid=1-s2.0-S0169500224004513-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906924","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}