Pub Date : 2024-09-30Epub Date: 2024-09-24DOI: 10.21037/tlcr-24-345
Dongsheng Wu, Jian Zhou, Lujia Song, Quan Zheng, Tengyong Wang, Zhizhen Ren, Yuchen Huang, Shuqiao Liu, Lunxu Liu
Background: Observational studies have revealed a potential association between gastroesophageal reflux disease (GERD) and lung cancer (LC), but the genetic role in their comorbidity have not been fully elucidated. This study aimed to comprehensively dissect the genetic link underlying GERD and LC.
Methods: Using large-scale genome-wide association study (GWAS) data, we investigated shared genetic architecture between GERD and LC. Our analyses encompassed genetic correlation, cross-trait meta-analysis, transcriptome-wide association studies (TWASs), and the evaluation of the causality though a bidirectional Mendelian randomization (MR) analysis with sufficient sensitivities.
Results: We identified a significant genome-wide genetic correlation between GERD and overall LC (rg =0.33, P=1.58×10-14), as well as across other subtype-specific LC (rg ranging from 0.19 to 0.39). After separating the whole genome into approximately 2,353 independent regions, 5 specific regions demonstrated significant local genetic correlation, with most significant region located at 9q33.3. Cross-trait meta-analysis revealed 22 pleiotropic loci between GERD and LC, including 3 novel loci (rs537160, rs10156445, and rs17391694). TWASs discovered a total of 49 genes shared in multiple tissues, such as lung tissues, esophagus muscularis, esophagus mucosa, and esophagus gastroesophageal junction. MR analysis suggested a significantly causal relationship between GERD and overall LC [odds ratio (OR) =1.34, 95% confidence interval (CI): 1.19-1.51], as well as other subtype-specific LC (OR ranging from 1.25 to 1.76). No evidence supports a significant causal effect of LC on GERD.
Conclusions: Our findings suggest intrinsic genetic correlation underlying GERD and LC, which provides valuable insights for screening and management of LC in individuals with GERD.
{"title":"A multi-level investigation of the genetic relationship between gastroesophageal reflux disease and lung cancer.","authors":"Dongsheng Wu, Jian Zhou, Lujia Song, Quan Zheng, Tengyong Wang, Zhizhen Ren, Yuchen Huang, Shuqiao Liu, Lunxu Liu","doi":"10.21037/tlcr-24-345","DOIUrl":"10.21037/tlcr-24-345","url":null,"abstract":"<p><strong>Background: </strong>Observational studies have revealed a potential association between gastroesophageal reflux disease (GERD) and lung cancer (LC), but the genetic role in their comorbidity have not been fully elucidated. This study aimed to comprehensively dissect the genetic link underlying GERD and LC.</p><p><strong>Methods: </strong>Using large-scale genome-wide association study (GWAS) data, we investigated shared genetic architecture between GERD and LC. Our analyses encompassed genetic correlation, cross-trait meta-analysis, transcriptome-wide association studies (TWASs), and the evaluation of the causality though a bidirectional Mendelian randomization (MR) analysis with sufficient sensitivities.</p><p><strong>Results: </strong>We identified a significant genome-wide genetic correlation between GERD and overall LC (<i>r<sub>g</sub></i> =0.33, P=1.58×10<sup>-14</sup>), as well as across other subtype-specific LC (<i>r<sub>g</sub></i> ranging from 0.19 to 0.39). After separating the whole genome into approximately 2,353 independent regions, 5 specific regions demonstrated significant local genetic correlation, with most significant region located at 9q33.3. Cross-trait meta-analysis revealed 22 pleiotropic loci between GERD and LC, including 3 novel loci (rs537160, rs10156445, and rs17391694). TWASs discovered a total of 49 genes shared in multiple tissues, such as lung tissues, esophagus muscularis, esophagus mucosa, and esophagus gastroesophageal junction. MR analysis suggested a significantly causal relationship between GERD and overall LC [odds ratio (OR) =1.34, 95% confidence interval (CI): 1.19-1.51], as well as other subtype-specific LC (OR ranging from 1.25 to 1.76). No evidence supports a significant causal effect of LC on GERD.</p><p><strong>Conclusions: </strong>Our findings suggest intrinsic genetic correlation underlying GERD and LC, which provides valuable insights for screening and management of LC in individuals with GERD.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484728/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Malignant pleural effusion (MPE) is associated with poor prognosis in patients with advanced lung adenocarcinoma (LUAD), and abnormal activation of epidermal growth factor receptor (EGFR) plays a crucial role in the development of LUAD. This study aimed to investigate the correlation between EGFR mutations and the occurrence of MPE in patients with LUAD and evaluate the effect of EGFR mutations on the prognosis of patients with LUAD with MPE.
Methods: A case-control study design was adopted that included patients pathologically diagnosed with LUAD. Clinical data were collected, and patients were divided into the MPE group and the non-MPE (N-MPE) group based on the presence of MPE. Propensity score matching (PSM) was used to control for confounding factors. The correlation between EGFR mutations and the occurrence of MPE in LUAD was initially examined. Additionally, various factors affecting the overall survival (OS) of patients with LUAD and MPE were evaluated.
Results: A total of 849 patients were included in the study. After 1:2 PSM, there were 180 patients in the MPE group and 360 in the N-MPE group. The EGFR mutation rate was significantly higher in the MPE group compared to the N-MPE group [62.7% vs. 50.2%; odds ratio (OR) =1.668; P=0.006]. This difference was primarily attributed to the T790M mutation (8.3% vs. 1.3%; OR =8.015; P<0.001), but no significant differences observed in other mutation sites between the groups. Further evaluation of factors affecting OS in patients with LUAD and MPE revealed that EGFR mutation was an independent protective factor for OS [hazard ratio (HR) 0.662, 95% CI: 0.456-0.962; P=0.03]. For patients with LUAD, MPE, and EGFR mutations, treatment with third-generation EGFR-tyrosine kinase inhibitors (TKIs) alone (HR 0.466, 95% CI: 0.233-0.930; P=0.03) or sequential first- and third-generation EGFR-TKIs (HR 0.385, 95% CI: 0.219-0.676; P=0.001) was associated with better median OS compared to first-generation EGFR-TKIs alone (first-generation EGFR-TKIs: 35 months, 95% CI: 28.4-41.6; third-generation EGFR-TKIs: 50 months, 95% CI: 37.3-62.7; sequential first- and third-generation EGFR-TKIs: 51 months, 95% CI: 45.6-56.4; P<0.001).
Conclusions: This study found there to be a positive correlation between EGFR mutations, particularly the T790M mutation, and MPE in patients with LUAD. EGFR mutation was associated with improved OS in patients with LUAD and MPE. For patients with LUAD, MPE, and EGFR mutations, sequential treatment with first- and third-generation EGFR-TKIs or third-generation EGFR-TKIs alone is recommended, as these regimens provide significant benefit to OS.
{"title":"<i>EGFR</i> mutations in patients with lung adenocarcinoma and malignant pleural effusion: a propensity score-matched analysis of a single-center database.","authors":"Qiwei Yang, Ziyi Wang, Qiang Fu, Xiaohai Hu, Liang Chen, Weiyang Chen, Ling Lv, Zhenghua Liu, Wanfu Men, Danni Li, Wenya Li","doi":"10.21037/tlcr-24-757","DOIUrl":"10.21037/tlcr-24-757","url":null,"abstract":"<p><strong>Background: </strong>Malignant pleural effusion (MPE) is associated with poor prognosis in patients with advanced lung adenocarcinoma (LUAD), and abnormal activation of epidermal growth factor receptor (EGFR) plays a crucial role in the development of LUAD. This study aimed to investigate the correlation between <i>EGFR</i> mutations and the occurrence of MPE in patients with LUAD and evaluate the effect of <i>EGFR</i> mutations on the prognosis of patients with LUAD with MPE.</p><p><strong>Methods: </strong>A case-control study design was adopted that included patients pathologically diagnosed with LUAD. Clinical data were collected, and patients were divided into the MPE group and the non-MPE (N-MPE) group based on the presence of MPE. Propensity score matching (PSM) was used to control for confounding factors. The correlation between <i>EGFR</i> mutations and the occurrence of MPE in LUAD was initially examined. Additionally, various factors affecting the overall survival (OS) of patients with LUAD and MPE were evaluated.</p><p><strong>Results: </strong>A total of 849 patients were included in the study. After 1:2 PSM, there were 180 patients in the MPE group and 360 in the N-MPE group. The <i>EGFR</i> mutation rate was significantly higher in the MPE group compared to the N-MPE group [62.7% <i>vs.</i> 50.2%; odds ratio (OR) =1.668; P=0.006]. This difference was primarily attributed to the T790M mutation (8.3% <i>vs.</i> 1.3%; OR =8.015; P<0.001), but no significant differences observed in other mutation sites between the groups. Further evaluation of factors affecting OS in patients with LUAD and MPE revealed that <i>EGFR</i> mutation was an independent protective factor for OS [hazard ratio (HR) 0.662, 95% CI: 0.456-0.962; P=0.03]. For patients with LUAD, MPE, and <i>EGFR</i> mutations, treatment with third-generation <i>EGFR</i>-tyrosine kinase inhibitors (TKIs) alone (HR 0.466, 95% CI: 0.233-0.930; P=0.03) or sequential first- and third-generation <i>EGFR</i>-TKIs (HR 0.385, 95% CI: 0.219-0.676; P=0.001) was associated with better median OS compared to first-generation <i>EGFR</i>-TKIs alone (first-generation <i>EGFR</i>-TKIs: 35 months, 95% CI: 28.4-41.6; third-generation <i>EGFR</i>-TKIs: 50 months, 95% CI: 37.3-62.7; sequential first- and third-generation <i>EGFR</i>-TKIs: 51 months, 95% CI: 45.6-56.4; P<0.001).</p><p><strong>Conclusions: </strong>This study found there to be a positive correlation between <i>EGFR</i> mutations, particularly the T790M mutation, and MPE in patients with LUAD. <i>EGFR</i> mutation was associated with improved OS in patients with LUAD and MPE. For patients with LUAD, MPE, and <i>EGFR</i> mutations, sequential treatment with first- and third-generation <i>EGFR</i>-TKIs or third-generation <i>EGFR</i>-TKIs alone is recommended, as these regimens provide significant benefit to OS.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484724/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475650","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 : 2024-09-30Epub Date: 2024-09-12DOI: 10.21037/tlcr-24-308
Per Hydbring
{"title":"Circulating tumor DNA (ctDNA)-the next generation biomarker in non-small cell lung cancer patients treated with immunotherapy?","authors":"Per Hydbring","doi":"10.21037/tlcr-24-308","DOIUrl":"10.21037/tlcr-24-308","url":null,"abstract":"","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484727/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475664","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 : 2024-09-30Epub Date: 2024-09-27DOI: 10.21037/tlcr-24-700
Zi-Ming Wang, Liang Guo, Yang Yang, Bo Tao, Wen-Qiang Zhang, Diego Gonzalez-Rivas, Jens-C Rueckert, Chee Yik Er, Calvin S H Ng, Moshe Lapidot, Gaetano Rocco, Mahmoud Ismail, Chen-Lu Yang, De-Ping Zhao
Background: Pneumonectomy is one of the important surgical methods for non-small cell lung cancer (NSCLC). This study evaluated the effects of laterality on the short- and long-term survival of NSCLC patients undergoing pneumonectomy.
Methods: We reviewed the Surveillance, Epidemiology, and End Results database to retrieve the data of patients who underwent pneumonectomy for stage I-III NSCLC from 2004 to 2015. Propensity score matching (PSM) was used to reduce the selection bias. Logistic regression was used to analyze the correlation between laterality and mortality at 3, 6, and 9 months. The Kaplan-Meier curve was used to further assess the effect of laterality on overall survival (OS).
Results: A total of 4,763 patients met the enrollment criteria [right-sided, 1,988 (41.7%); left-sided, 2,775 (58.3%)]. After PSM, 1,911 patients for each side were included in the further analysis. The first 6 months following pneumonectomy was the main period of death, with 32.0% (428/1,336) and 19.9% (250/1,258) of right- and left-sided deaths occurring during this period. The logistic regression analysis showed that right-sided pneumonectomy was an independent risk factor for 3- (P<0.001) and 6-month (P<0.001) mortality. However, laterality had no significant effect on postoperative death at 7-9 months (P=0.82). In the total cohort, right-sided patients had worse OS (P<0.001), but the subgroup survival analysis of patients with a follow-up period >6 months revealed that laterality had no statistically significant effect on OS (P=0.75).
Conclusions: Right-sided pneumonectomy was associated with a higher perioperative mortality risk that lasted about 6 months. After that period, laterality was not observed to have a significant prognostic effect on the OS of patients undergoing pneumonectomy.
{"title":"Effect of laterality on the postoperative survival of non-small cell lung cancer patients undergoing pneumonectomy.","authors":"Zi-Ming Wang, Liang Guo, Yang Yang, Bo Tao, Wen-Qiang Zhang, Diego Gonzalez-Rivas, Jens-C Rueckert, Chee Yik Er, Calvin S H Ng, Moshe Lapidot, Gaetano Rocco, Mahmoud Ismail, Chen-Lu Yang, De-Ping Zhao","doi":"10.21037/tlcr-24-700","DOIUrl":"10.21037/tlcr-24-700","url":null,"abstract":"<p><strong>Background: </strong>Pneumonectomy is one of the important surgical methods for non-small cell lung cancer (NSCLC). This study evaluated the effects of laterality on the short- and long-term survival of NSCLC patients undergoing pneumonectomy.</p><p><strong>Methods: </strong>We reviewed the Surveillance, Epidemiology, and End Results database to retrieve the data of patients who underwent pneumonectomy for stage I-III NSCLC from 2004 to 2015. Propensity score matching (PSM) was used to reduce the selection bias. Logistic regression was used to analyze the correlation between laterality and mortality at 3, 6, and 9 months. The Kaplan-Meier curve was used to further assess the effect of laterality on overall survival (OS).</p><p><strong>Results: </strong>A total of 4,763 patients met the enrollment criteria [right-sided, 1,988 (41.7%); left-sided, 2,775 (58.3%)]. After PSM, 1,911 patients for each side were included in the further analysis. The first 6 months following pneumonectomy was the main period of death, with 32.0% (428/1,336) and 19.9% (250/1,258) of right- and left-sided deaths occurring during this period. The logistic regression analysis showed that right-sided pneumonectomy was an independent risk factor for 3- (P<0.001) and 6-month (P<0.001) mortality. However, laterality had no significant effect on postoperative death at 7-9 months (P=0.82). In the total cohort, right-sided patients had worse OS (P<0.001), but the subgroup survival analysis of patients with a follow-up period >6 months revealed that laterality had no statistically significant effect on OS (P=0.75).</p><p><strong>Conclusions: </strong>Right-sided pneumonectomy was associated with a higher perioperative mortality risk that lasted about 6 months. After that period, laterality was not observed to have a significant prognostic effect on the OS of patients undergoing pneumonectomy.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484732/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Lung cancer was often diagnosed by malignant pleural effusion (MPE). Excessive MPE is generally discarded. The establishment of cell lines and the generation of cancer mouse models have the potential to be directly linked to personalized medicine. This study aimed to establish cell lines and generate mouse models using MPE.
Methods: Cells derived from 5 mL of MPE were cultured in several conditions, including 100% MPE supernatant and Roswell Park Memorial Institute-1640 supplemented with 10% fetal bovine serum (FBS) or 10% MPE supernatant. When steady cell growth was observed, fewer cells were spread and the colonies were selected to establish the cell line. Cells derived from 10 mL of MPE were inoculated subcutaneously into non-obese diabetic-severe combined immunodeficiency (NOD-scid) and NOD.Cg-Prkdcscid Il2rgtmlWjl /SzJ (NSG) mice to assess tumorigenic potential.
Results: MPEs were obtained from 28 lung cancer patients, 23 of whom had adenocarcinoma. Cell lines were established from 5 patients (18%). Tumorigenesis was observed in 6 of 28 cases (21%). However, in 7 cases, the mice (7 NSG and 1 NOD-scid mice) became progressively weaker, lost their hair, and died within 12 weeks without tumorigenesis. The appearance and pathological findings were consistent with graft-versus-host disease. Cell line establishment and tumorigenesis in mice were associated with a lower response to first-line therapy and poorer prognosis of patients.
Conclusions: When MPEs were simply utilized, the cell line establishment rate was 18% and the engraftment rate in mice was 21%. The prognosis of patients who underwent cell line establishment and engraftment in mice was poor.
{"title":"Establishment of lung cancer cell lines and tumorigenesis in mice from malignant pleural effusion in patients with lung cancer.","authors":"Nobuhiro Kanaji, Masanao Yokohira, Takuya Inoue, Naoki Watanabe, Hitoshi Mizoguchi, Yuta Komori, Kosuke Kawada, Norimitsu Kadowaki","doi":"10.21037/tlcr-24-143","DOIUrl":"10.21037/tlcr-24-143","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer was often diagnosed by malignant pleural effusion (MPE). Excessive MPE is generally discarded. The establishment of cell lines and the generation of cancer mouse models have the potential to be directly linked to personalized medicine. This study aimed to establish cell lines and generate mouse models using MPE.</p><p><strong>Methods: </strong>Cells derived from 5 mL of MPE were cultured in several conditions, including 100% MPE supernatant and Roswell Park Memorial Institute-1640 supplemented with 10% fetal bovine serum (FBS) or 10% MPE supernatant. When steady cell growth was observed, fewer cells were spread and the colonies were selected to establish the cell line. Cells derived from 10 mL of MPE were inoculated subcutaneously into non-obese diabetic-severe combined immunodeficiency (NOD-<i>scid</i>) and NOD.Cg-<i>Prkdc<sup>scid</sup> Il2rg<sup>tmlWjl</sup></i> /SzJ (NSG) mice to assess tumorigenic potential.</p><p><strong>Results: </strong>MPEs were obtained from 28 lung cancer patients, 23 of whom had adenocarcinoma. Cell lines were established from 5 patients (18%). Tumorigenesis was observed in 6 of 28 cases (21%). However, in 7 cases, the mice (7 NSG and 1 NOD-<i>scid</i> mice) became progressively weaker, lost their hair, and died within 12 weeks without tumorigenesis. The appearance and pathological findings were consistent with graft-versus-host disease. Cell line establishment and tumorigenesis in mice were associated with a lower response to first-line therapy and poorer prognosis of patients.</p><p><strong>Conclusions: </strong>When MPEs were simply utilized, the cell line establishment rate was 18% and the engraftment rate in mice was 21%. The prognosis of patients who underwent cell line establishment and engraftment in mice was poor.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484711/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475680","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 : 2024-09-30Epub Date: 2024-09-21DOI: 10.21037/tlcr-24-241
George Corby, Nicola L Barclay, Eng Hooi Tan, Edward Burn, Antonella Delmestri, Talita Duarte-Salles, Asieh Golozar, Wai Yi Man, Ilona Tietzova, Daniel Prieto-Alhambra, Danielle Newby
Background: Lung cancer is the leading cause of cancer-associated mortality worldwide. In the United Kingdom (UK), there has been a major reduction in smoking, the leading risk factor for lung cancer. Therefore, an up-to-date assessment of the trends of lung cancer is required in the UK. This study aims to describe lung cancer burden and trends in terms of incidence, prevalence, and survival from 2000-2021, using two UK primary care databases.
Methods: We performed a population-based cohort study using the UK primary care Clinical Practice Research Datalink (CPRD) GOLD database, compared with CPRD Aurum. Participants aged 18+ years, with 1-year of prior data availability, were included. We estimated lung cancer incidence rates (IRs), period prevalence (PP), and survival at 1, 5 and 10 years after diagnosis using the Kaplan-Meier (KM) method.
Results: Overall, 11,388,117 participants, with 45,563 lung cancer cases were studied. The IR of lung cancer was 52.0 [95% confidence interval (CI): 51.5 to 52.5] per 100,000 person-years, with incidence increasing from 2000 to 2021. Females aged over 50 years of age showed increases in incidence over the study period, ranging from increases of 8 to 123 per 100,000 person-years, with the greatest increase in females aged 80-89 years. Alternatively, for males, only cohorts aged over 80 years showed increases in incidence over the study period. The highest IR was observed in people aged 80-89 years. PP in 2021 was 0.18%, with the largest rise seen in participants aged over 60 years. Median survival post-diagnosis increased from 6.6 months in those diagnosed between 2000-2004 to 10.0 months between 2015-2019. Both short and long-term survival was higher in younger cohorts, with 82.7% 1-year survival in those aged 18-29 years, versus 24.2% in the age 90+ years cohort. Throughout the study period, survival was longer in females, with a larger increase in survival over time than in males.
Conclusions: The incidence and prevalence of lung cancer diagnoses in the UK have increased, especially in female and older populations, with a small increase in median survival. This study will enable future comparisons of overall disease burden, so the overall impact may be seen.
{"title":"Incidence, prevalence, and survival of lung cancer in the United Kingdom from 2000-2021: a population-based cohort study.","authors":"George Corby, Nicola L Barclay, Eng Hooi Tan, Edward Burn, Antonella Delmestri, Talita Duarte-Salles, Asieh Golozar, Wai Yi Man, Ilona Tietzova, Daniel Prieto-Alhambra, Danielle Newby","doi":"10.21037/tlcr-24-241","DOIUrl":"10.21037/tlcr-24-241","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is the leading cause of cancer-associated mortality worldwide. In the United Kingdom (UK), there has been a major reduction in smoking, the leading risk factor for lung cancer. Therefore, an up-to-date assessment of the trends of lung cancer is required in the UK. This study aims to describe lung cancer burden and trends in terms of incidence, prevalence, and survival from 2000-2021, using two UK primary care databases.</p><p><strong>Methods: </strong>We performed a population-based cohort study using the UK primary care Clinical Practice Research Datalink (CPRD) GOLD database, compared with CPRD Aurum. Participants aged 18+ years, with 1-year of prior data availability, were included. We estimated lung cancer incidence rates (IRs), period prevalence (PP), and survival at 1, 5 and 10 years after diagnosis using the Kaplan-Meier (KM) method.</p><p><strong>Results: </strong>Overall, 11,388,117 participants, with 45,563 lung cancer cases were studied. The IR of lung cancer was 52.0 [95% confidence interval (CI): 51.5 to 52.5] per 100,000 person-years, with incidence increasing from 2000 to 2021. Females aged over 50 years of age showed increases in incidence over the study period, ranging from increases of 8 to 123 per 100,000 person-years, with the greatest increase in females aged 80-89 years. Alternatively, for males, only cohorts aged over 80 years showed increases in incidence over the study period. The highest IR was observed in people aged 80-89 years. PP in 2021 was 0.18%, with the largest rise seen in participants aged over 60 years. Median survival post-diagnosis increased from 6.6 months in those diagnosed between 2000-2004 to 10.0 months between 2015-2019. Both short and long-term survival was higher in younger cohorts, with 82.7% 1-year survival in those aged 18-29 years, versus 24.2% in the age 90+ years cohort. Throughout the study period, survival was longer in females, with a larger increase in survival over time than in males.</p><p><strong>Conclusions: </strong>The incidence and prevalence of lung cancer diagnoses in the UK have increased, especially in female and older populations, with a small increase in median survival. This study will enable future comparisons of overall disease burden, so the overall impact may be seen.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484731/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475683","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Lung large cell neuroendocrine carcinoma (LCNEC) is an aggressive disease with poor prognosis and short-term survival, which lacks effective prognostic indicators. The study aims to investigate the molecular subtypes and prognostic markers of lung LCNEC.
Methods: Patients diagnosed with lung LCNEC at Sun Yat-sen University Cancer Center (SYSUCC) between November 2007 and January 2021 were screened. Baseline clinical data were collected and routine blood indexes including lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammation index (SII) were calculated. Immunohistochemistry (IHC) of ASCL1, NEUROD1, POU2F3, YAP1 were done to perform molecular subtyping, while CD56, Syn, CgA, CD3, CD8, CD20, CD68, and CD163 were also stained on tissue samples. Then prognostic factors of lung LCNEC were explored.
Results: One hundred and fifty-one lung LCNEC patients were identified, 103 of whom had complete clinical information, available routine blood and biochemical indexes were eventually included in the present study. Tumor tissue specimens were available from 64 patients. Positive expression rates of ASCL1, NEUROD1, and YAP1 were 82.8%, 50.0%, and 28.1%, respectively. No POU2F3+ cases were detected. Forty (62.5%) patients co-expressed with two or three markers. High LMR (>3.3) was an independent predictor of favorable prognosis of disease-free survival (DFS) [hazard ratio (HR), 0.391; 95% confidence interval (CI): 0.161-0.948; P=0.04] and overall survival (OS) (HR, 0.201; 95% CI: 0.071-0.574; P=0.003). Notably, high LMR was correlated with higher intra-tumoral CD3+ (P=0.004), CD8+ (P=0.01), and CD68+ (P<0.001) immune cell infiltration compared to low LMR in lung LCNEC.
Conclusions: Our study validated molecular subtypes by IHC in lung LCNEC, and co-expression was found among different subtypes, with no prognostic effect. High blood LMR level was associated with a favorable prognosis in lung LCNEC, which might partly reflect a hot tumor tissue immune microenvironment. Our findings may benefit clinical practice, and further studies are warranted.
{"title":"Molecular subtypes and prognostic factors of lung large cell neuroendocrine carcinoma.","authors":"Tingting Liu, Xueyuan Chen, Silang Mo, Ting Zhou, Wenjuan Ma, Gang Chen, Xiang Chen, Mengting Shi, Yuwen Yang, Yan Huang, Hongyun Zhao, Wenfeng Fang, Yunpeng Yang, Jing Li, Li Zhang, Yuanyuan Zhao","doi":"10.21037/tlcr-24-292","DOIUrl":"10.21037/tlcr-24-292","url":null,"abstract":"<p><strong>Background: </strong>Lung large cell neuroendocrine carcinoma (LCNEC) is an aggressive disease with poor prognosis and short-term survival, which lacks effective prognostic indicators. The study aims to investigate the molecular subtypes and prognostic markers of lung LCNEC.</p><p><strong>Methods: </strong>Patients diagnosed with lung LCNEC at Sun Yat-sen University Cancer Center (SYSUCC) between November 2007 and January 2021 were screened. Baseline clinical data were collected and routine blood indexes including lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammation index (SII) were calculated. Immunohistochemistry (IHC) of ASCL1, NEUROD1, POU2F3, YAP1 were done to perform molecular subtyping, while CD56, Syn, CgA, CD3, CD8, CD20, CD68, and CD163 were also stained on tissue samples. Then prognostic factors of lung LCNEC were explored.</p><p><strong>Results: </strong>One hundred and fifty-one lung LCNEC patients were identified, 103 of whom had complete clinical information, available routine blood and biochemical indexes were eventually included in the present study. Tumor tissue specimens were available from 64 patients. Positive expression rates of ASCL1, NEUROD1, and YAP1 were 82.8%, 50.0%, and 28.1%, respectively. No POU2F3<sup>+</sup> cases were detected. Forty (62.5%) patients co-expressed with two or three markers. High LMR (>3.3) was an independent predictor of favorable prognosis of disease-free survival (DFS) [hazard ratio (HR), 0.391; 95% confidence interval (CI): 0.161-0.948; P=0.04] and overall survival (OS) (HR, 0.201; 95% CI: 0.071-0.574; P=0.003). Notably, high LMR was correlated with higher intra-tumoral CD3<sup>+</sup> (P=0.004), CD8<sup>+</sup> (P=0.01), and CD68<sup>+</sup> (P<0.001) immune cell infiltration compared to low LMR in lung LCNEC.</p><p><strong>Conclusions: </strong>Our study validated molecular subtypes by IHC in lung LCNEC, and co-expression was found among different subtypes, with no prognostic effect. High blood LMR level was associated with a favorable prognosis in lung LCNEC, which might partly reflect a hot tumor tissue immune microenvironment. Our findings may benefit clinical practice, and further studies are warranted.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484736/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475685","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 : 2024-09-30Epub Date: 2024-09-27DOI: 10.21037/tlcr-24-284
Linda Ye, Heeju Ryu, David Granadier, Long T Nguyen, Yannick Simoni, Ian Dick, Tina Firth, Ebony Rouse, Peter Chiang, Y C Gary Lee, Bruce W Robinson, Jenette Creaney, Evan W Newell, Alec J Redwood
Background: Anti-tumor CD8 T cells are important for immunity but can become 'exhausted' and hence ineffective. Tumor-infiltrating exhausted CD8+ T cells include less differentiated stem-like exhausted T (Texstem) cells and terminally exhausted T (Texterm) cells. Both subsets have been proposed as prognostic biomarkers in cancer patients. In this study, we retrospectively investigated their prognostic significance in patients with metastatic non-small cell lung cancer (NSCLC) and validated our findings in a mesothelioma cohort.
Methods: Pre-treatment malignant pleural effusions (PEs) from 43 NSCLC (41 non-squamous, 2 squamous) patients were analyzed by flow cytometry. The percentages of Texstem and Texterm CD8 T cells were correlated with overall survival (OS) after adjusting for clinicopathological variables. Findings were validated using a mesothelioma cohort (n=49). Mass cytometry was performed on 16 pre-treatment PE samples from 5 mesothelioma and 3 NSCLC patients for T-cell phenotyping. Single-cell multi-omics analysis was performed on 4 pre-treatment PE samples from 2 NSCLC patients and 2 mesothelioma patients for analysis of the transcriptomic profiles, surface markers and T cell receptor (TCR) repertoire.
Results: Higher frequency of Texstem was associated with significantly increased OS [median 9.9 vs. 3.4 months, hazard ratio (HR) 0.36, 95% CI: 0.16-0.79, P=0.01]. The frequency of Texterm was not associated with OS. These findings were validated in the mesothelioma cohort (high vs. low Texstem, median OS 32.1 vs. 19.8 months, HR 0.31, 95% CI: 0.10-0.96, P=0.04). Detailed single-cell sequencing and mass cytometry profiling revealed that exhausted T cells from NSCLC expressed greater stem-likeness and less inhibitory markers than those from mesothelioma and that Texstem cells also contained 'bystander' virus-specific T cells.
Conclusions: This study demonstrates that PE CD8 Texstem cell abundance is associated with better survival outcomes, and thus may be a useful prognostic biomarker.
背景:抗肿瘤 CD8 T 细胞对免疫非常重要,但也会 "衰竭",从而失去作用。肿瘤浸润的衰竭 CD8+ T 细胞包括分化程度较低的干样衰竭 T 细胞(Texstem)和终末衰竭 T 细胞(Texterm)。这两个亚群被认为是癌症患者的预后生物标志物。在这项研究中,我们回顾性地调查了它们在转移性非小细胞肺癌(NSCLC)患者中的预后意义,并在间皮瘤队列中验证了我们的发现:方法:采用流式细胞术分析了43例NSCLC患者(41例非鳞癌,2例鳞癌)治疗前的恶性胸腔积液(PE)。在调整临床病理变量后,Texstem 和 Texterm CD8 T 细胞的百分比与总生存率(OS)相关。研究结果通过间皮瘤队列(n=49)进行了验证。对来自 5 名间皮瘤患者和 3 名 NSCLC 患者的 16 份治疗前 PE 样本进行了 T 细胞表型分析,并使用了质谱细胞计数法。对来自 2 名 NSCLC 患者和 2 名间皮瘤患者的 4 份治疗前 PE 样本进行了单细胞多组学分析,以分析转录组图谱、表面标记和 T 细胞受体(TCR)谱系:Texstem频率越高,OS显著增加[中位数9.9个月对3.4个月,危险比(HR)0.36,95% CI:0.16-0.79,P=0.01]。Texterm的频率与OS无关。这些发现在间皮瘤队列中得到了验证(高Texterm与低Texterm,中位OS为32.1个月与19.8个月,HR为0.31,95% CI:0.10-0.96,P=0.04)。详细的单细胞测序和质谱分析显示,与间皮瘤的T细胞相比,NSCLC的衰竭T细胞表达了更多的干相似性和更少的抑制性标记,Texstem细胞还包含 "旁观者 "病毒特异性T细胞:这项研究表明,PE CD8 Texstem 细胞的丰度与较好的生存结果相关,因此可能是一种有用的预后生物标志物。
{"title":"Stem-like exhausted CD8 T cells in pleural effusions predict improved survival in non-small cell lung cancer (NSCLC) and mesothelioma.","authors":"Linda Ye, Heeju Ryu, David Granadier, Long T Nguyen, Yannick Simoni, Ian Dick, Tina Firth, Ebony Rouse, Peter Chiang, Y C Gary Lee, Bruce W Robinson, Jenette Creaney, Evan W Newell, Alec J Redwood","doi":"10.21037/tlcr-24-284","DOIUrl":"10.21037/tlcr-24-284","url":null,"abstract":"<p><strong>Background: </strong>Anti-tumor CD8 T cells are important for immunity but can become 'exhausted' and hence ineffective. Tumor-infiltrating exhausted CD8<sup>+</sup> T cells include less differentiated stem-like exhausted T (Tex<sup>stem</sup>) cells and terminally exhausted T (Tex<sup>term</sup>) cells. Both subsets have been proposed as prognostic biomarkers in cancer patients. In this study, we retrospectively investigated their prognostic significance in patients with metastatic non-small cell lung cancer (NSCLC) and validated our findings in a mesothelioma cohort.</p><p><strong>Methods: </strong>Pre-treatment malignant pleural effusions (PEs) from 43 NSCLC (41 non-squamous, 2 squamous) patients were analyzed by flow cytometry. The percentages of Tex<sup>stem</sup> and Tex<sup>term</sup> CD8 T cells were correlated with overall survival (OS) after adjusting for clinicopathological variables. Findings were validated using a mesothelioma cohort (n=49). Mass cytometry was performed on 16 pre-treatment PE samples from 5 mesothelioma and 3 NSCLC patients for T-cell phenotyping. Single-cell multi-omics analysis was performed on 4 pre-treatment PE samples from 2 NSCLC patients and 2 mesothelioma patients for analysis of the transcriptomic profiles, surface markers and T cell receptor (TCR) repertoire.</p><p><strong>Results: </strong>Higher frequency of Tex<sup>stem</sup> was associated with significantly increased OS [median 9.9 <i>vs.</i> 3.4 months, hazard ratio (HR) 0.36, 95% CI: 0.16-0.79, P=0.01]. The frequency of Tex<sup>term</sup> was not associated with OS. These findings were validated in the mesothelioma cohort (high <i>vs.</i> low Tex<sup>stem</sup>, median OS 32.1 <i>vs.</i> 19.8 months, HR 0.31, 95% CI: 0.10-0.96, P=0.04). Detailed single-cell sequencing and mass cytometry profiling revealed that exhausted T cells from NSCLC expressed greater stem-likeness and less inhibitory markers than those from mesothelioma and that Tex<sup>stem</sup> cells also contained 'bystander' virus-specific T cells.</p><p><strong>Conclusions: </strong>This study demonstrates that PE CD8 Tex<sup>stem</sup> cell abundance is associated with better survival outcomes, and thus may be a useful prognostic biomarker.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484714/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475698","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 : 2024-09-30Epub Date: 2024-09-24DOI: 10.21037/tlcr-24-157
Yan Hu, Chao Zeng, Jina Li, Siying Ren, Mengqi Shao, Weixuan Lei, Junqi Yi, Wei Han, Jieming Cao, Jian Zou, Quanming Fei, Zeyu Cheng, Wenliang Liu
Background: Efficient ground-glass opacities (GGOs) diagnosis is challenging. A diagnostic method distinguishing malignant from benign GGOs is warranted. In this study, we sought to construct a noninvasive method based on tumor educated platelet (TEP) RNA profiles for malignant GGOs diagnosis and explore the molecular mechanism of the potential biomarker for the first time.
Methods: Based on TEP RNA-sequencing (TEP RNA-seq) in benign and malignant GGOs, a classification model was constructed using differentially expressed genes (DEGs) and was used to evaluate diagnostic performance. High-throughput quantitative polymerase chain reaction (HT-qPCR) verified 23 genes selected from the top 60 DEGs between benign and malignant GGOs. The correlation between 17 verified DEGs and 22 key glycolytic genes was analyzed. Tripartite motif-containing 27 (TRIM27) overexpressing and knockdown (KD) cell models were constructed using A549 and PC-9 cells, respectively in which cell growth, apoptosis, migration and invasion were evaluated. The protein levels of HK-1/2, PKM1/2, LDHA and GLUT1 were evaluated by western blot. Glycolysis was evaluated through adenosine triphosphate (ATP), reactive oxygen species (ROS), lactate acid (LD) production, glucose uptake, and lactate dehydrogenase (LDH) activity assays. RNA-seq was performed in loss-of TRIM27-KD PC-9 cells to clarify the downstream factors of TRIM27 which was verified using western blot and immunofluorescence double staining.
Results: In 81 samples, the 1,647-DEG-based classification model exhibited area under the curve (AUC), sensitivity, and specificity values of 0.99 [95% confidence interval (CI): 0.972-1.000], 100%, and 91%, respectively, while the top 60-DEG-based classification model exhibited AUC, sensitivity, and specificity values of 0.986 (95% CI: 0.962-1.000), 98%, and 91%, respectively. TRIM27 achieved AUC of 0.87 in the diagnosis of malignant GGOs, with 83.93% sensitivity, 78.79% specificity, 81.15% accuracy, 77.05% positive predictive value (PPV) and 85.25% negative predictive value (NPV). TRIM27 was highly expressed in non-small cell lung cancer (NSCLC) cells, and accelerated cell migration and invasion. In addition, TRIM27 was found to promote glycolysis in NSCLC cells partially through HMOX1 which was negatively correlated with TRIM27.
Conclusions: We constructed a novel TEP RNA-seq based classifier for malignant GGOs diagnosis. TRIM27, an important target discovered, could accelerate migration, invasion and regulate glycolysis partially through HMOX1 in NSCLC cells, thus providing scientific support for TRIM27 as a diagnostic biomarker for malignant GGO diagnosis.
{"title":"TRIM27 revealing by tumor educated platelet RNA-sequencing, as a potential biomarker for malignant ground-glass opacities diagnosis mediates glycolysis of non-small cell lung cancer cells partially through HOXM1.","authors":"Yan Hu, Chao Zeng, Jina Li, Siying Ren, Mengqi Shao, Weixuan Lei, Junqi Yi, Wei Han, Jieming Cao, Jian Zou, Quanming Fei, Zeyu Cheng, Wenliang Liu","doi":"10.21037/tlcr-24-157","DOIUrl":"10.21037/tlcr-24-157","url":null,"abstract":"<p><strong>Background: </strong>Efficient ground-glass opacities (GGOs) diagnosis is challenging. A diagnostic method distinguishing malignant from benign GGOs is warranted. In this study, we sought to construct a noninvasive method based on tumor educated platelet (TEP) RNA profiles for malignant GGOs diagnosis and explore the molecular mechanism of the potential biomarker for the first time.</p><p><strong>Methods: </strong>Based on TEP RNA-sequencing (TEP RNA-seq) in benign and malignant GGOs, a classification model was constructed using differentially expressed genes (DEGs) and was used to evaluate diagnostic performance. High-throughput quantitative polymerase chain reaction (HT-qPCR) verified 23 genes selected from the top 60 DEGs between benign and malignant GGOs. The correlation between 17 verified DEGs and 22 key glycolytic genes was analyzed. Tripartite motif-containing 27 (TRIM27) overexpressing and knockdown (KD) cell models were constructed using A549 and PC-9 cells, respectively in which cell growth, apoptosis, migration and invasion were evaluated. The protein levels of HK-1/2, PKM1/2, LDHA and GLUT1 were evaluated by western blot. Glycolysis was evaluated through adenosine triphosphate (ATP), reactive oxygen species (ROS), lactate acid (LD) production, glucose uptake, and lactate dehydrogenase (LDH) activity assays. RNA-seq was performed in loss-of TRIM27-KD PC-9 cells to clarify the downstream factors of TRIM27 which was verified using western blot and immunofluorescence double staining.</p><p><strong>Results: </strong>In 81 samples, the 1,647-DEG-based classification model exhibited area under the curve (AUC), sensitivity, and specificity values of 0.99 [95% confidence interval (CI): 0.972-1.000], 100%, and 91%, respectively, while the top 60-DEG-based classification model exhibited AUC, sensitivity, and specificity values of 0.986 (95% CI: 0.962-1.000), 98%, and 91%, respectively. TRIM27 achieved AUC of 0.87 in the diagnosis of malignant GGOs, with 83.93% sensitivity, 78.79% specificity, 81.15% accuracy, 77.05% positive predictive value (PPV) and 85.25% negative predictive value (NPV). TRIM27 was highly expressed in non-small cell lung cancer (NSCLC) cells, and accelerated cell migration and invasion. In addition, TRIM27 was found to promote glycolysis in NSCLC cells partially through HMOX1 which was negatively correlated with TRIM27.</p><p><strong>Conclusions: </strong>We constructed a novel TEP RNA-seq based classifier for malignant GGOs diagnosis. TRIM27, an important target discovered, could accelerate migration, invasion and regulate glycolysis partially through HMOX1 in NSCLC cells, thus providing scientific support for TRIM27 as a diagnostic biomarker for malignant GGO diagnosis.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484725/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475702","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 : 2024-09-30Epub Date: 2024-09-26DOI: 10.21037/tlcr-24-439
Edyta M Urbanska, Morten Grauslund, Siv M S Berger, Junia C Costa, Peter R Koffeldt, Jens B Sørensen, Eric Santoni-Rugiu
Background: Most patients with advanced anaplastic lymphoma kinase (ALK)-rearranged (ALK+) non-small cell lung cancer (NSCLC) experience prolonged response to second-generation (2G) ALK-tyrosine kinase inhibitors (TKIs). Herein, we present a case of metastatic ALK+ NSCLC rapidly progressing on first-line treatment due to de novo amplification of the mesenchymal-epithelial transition factor (MET) gene, which is a still elusive and underrecognized mechanism of primary resistance to ALK-TKIs.
Case description: A 43-year-old, female diagnosed with T4N3M1c NSCLC harboring the echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusion variant 1 (EML4-ALK v.1) and TP53 co-mutation, displayed only mixed response after three months and highly symptomatic progression after 6 months of first-line brigatinib treatment. Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) analysis on re-biopsies from a new liver metastasis revealed overexpression of MET receptor (3+ in 80% of tumor cells) and heterogeneously increased MET gene copy number (CN) in tumor cells, including 20% with MET clusters (corresponds to ≥15 gene copies, thus exact CN uncountable by FISH) and the other 80% with median MET CN of 8.3, both changes indicating high-level MET-amplification. DNA and RNA next-generation sequencing (NGS) displayed preserved ALK fusion and TP53 co-mutation, but no additional genomic alterations, nor MET-amplification. Therefore, we retrospectively investigated the diagnostic biopsy from the primary tumor in the left lung with IHC and FISH revealing the presence of increased MET receptor expression (2+ in 100% of tumor cells) and MET-amplification (median MET CN of 6.1), which otherwise was not detected by NGS. Thus, given the well-documented efficacy of alectinib towards EML4-ALK v.1, combined second-line treatment with alectinib and the MET-TKI, crizotinib, was implemented resulting in very pronounced objective response, significantly improved quality of life, and no adverse events so far during the ongoing treatment (6 months).
Conclusions: The combination of alectinib and crizotinib may be a feasible and effective treatment for ALK+ NSCLC with de novoMET-amplification. The latter may represent a mechanism of intrinsic ALK-TKI resistance and its recognition by FISH, in NGS-negative cases, may be considered before initiating first-line treatment. This recognition is clinically important as combined therapy with ALK-TKI and MET-inhibitor should be the preferred first-line treatment.
背景:大多数晚期无性淋巴瘤激酶(ALK)重排(ALK+)非小细胞肺癌(NSCLC)患者对第二代(2G)ALK-酪氨酸激酶抑制剂(TKIs)的反应延长。在此,我们介绍了一例因间质上皮转化因子(MET)基因的新生扩增而导致一线治疗迅速进展的转移性ALK+ NSCLC病例,MET基因扩增是ALK-TKIs原发性耐药的一种仍难以捉摸且未得到充分认识的机制:43岁女性,诊断为T4N3M1c NSCLC,携带棘皮微管相关蛋白样4(EML4)-ALK融合变体1(EML4-ALK v.1)和TP53共突变,三个月后仅出现混合反应,一线治疗6个月后出现高度症状性进展。免疫组化(IHC)和荧光原位杂交(FISH)分析显示,新发肝转移灶的活检组织中MET受体过表达(80%的肿瘤细胞3+),肿瘤细胞中MET基因拷贝数(CN)异质性增加,其中20%的肿瘤细胞中存在MET簇(相当于≥15个基因拷贝,因此FISH无法计算确切的CN),另外80%的肿瘤细胞中MET CN中位数为8.3,这两种变化均表明肿瘤细胞中存在高水平的MET扩增。DNA和RNA下一代测序(NGS)显示保留了ALK融合和TP53共突变,但没有其他基因组改变,也没有MET扩增。因此,我们通过 IHC 和 FISH 对左肺原发肿瘤的诊断性活检进行了回顾性研究,结果显示存在 MET 受体表达增加(100% 的肿瘤细胞为 2+)和 MET 扩增(中位数 MET CN 为 6.1),而 NGS 则未检测到这一情况。因此,考虑到阿来替尼对EML4-ALK v.1的疗效已得到充分证实,患者接受了阿来替尼和MET-TKI克唑替尼的联合二线治疗,结果获得了非常明显的客观反应,生活质量明显改善,并且在持续治疗期间(6个月)至今未出现任何不良反应:结论:阿来替尼和克唑替尼联用可能是一种可行且有效的治疗方法,适用于新发MET扩增的ALK+ NSCLC。后者可能代表了ALK-TKI的内在耐药机制,在NGS阴性病例中,可在开始一线治疗前考虑通过FISH识别其耐药性。这种识别在临床上非常重要,因为 ALK-TKI 和 MET 抑制剂的联合治疗应该是首选的一线治疗方法。
{"title":"ALK-tyrosine kinase inhibitor intrinsic resistance due to <i>de novo</i> <i>MET</i>-amplification in metastatic <i>ALK</i>-rearranged non-small cell lung cancer effectively treated by alectinib-crizotinib combination-case report.","authors":"Edyta M Urbanska, Morten Grauslund, Siv M S Berger, Junia C Costa, Peter R Koffeldt, Jens B Sørensen, Eric Santoni-Rugiu","doi":"10.21037/tlcr-24-439","DOIUrl":"10.21037/tlcr-24-439","url":null,"abstract":"<p><strong>Background: </strong>Most patients with advanced anaplastic lymphoma kinase (<i>ALK</i>)-rearranged (<i>ALK</i>+) non-small cell lung cancer (NSCLC) experience prolonged response to second-generation (2G) ALK-tyrosine kinase inhibitors (TKIs). Herein, we present a case of metastatic <i>ALK</i>+ NSCLC rapidly progressing on first-line treatment due to <i>de novo</i> amplification of the mesenchymal-epithelial transition factor (<i>MET</i>) gene, which is a still elusive and underrecognized mechanism of primary resistance to ALK-TKIs.</p><p><strong>Case description: </strong>A 43-year-old, female diagnosed with T4N3M1c NSCLC harboring the echinoderm microtubule-associated protein-like 4 (<i>EML4</i>)<i>-ALK</i> fusion variant 1 (<i>EML4-ALK</i> v.1) and <i>TP53</i> co-mutation, displayed only mixed response after three months and highly symptomatic progression after 6 months of first-line brigatinib treatment. Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) analysis on re-biopsies from a new liver metastasis revealed overexpression of MET receptor (3+ in 80% of tumor cells) and heterogeneously increased <i>MET</i> gene copy number (CN) in tumor cells, including 20% with <i>MET</i> clusters (corresponds to ≥15 gene copies, thus exact CN uncountable by FISH) and the other 80% with median <i>MET</i> CN of 8.3, both changes indicating high-level <i>MET-</i>amplification. DNA and RNA next-generation sequencing (NGS) displayed preserved <i>ALK</i> fusion and <i>TP53</i> co-mutation, but no additional genomic alterations, nor <i>MET-</i>amplification. Therefore, we retrospectively investigated the diagnostic biopsy from the primary tumor in the left lung with IHC and FISH revealing the presence of increased MET receptor expression (2+ in 100% of tumor cells) and <i>MET-</i>amplification (median <i>MET</i> CN of 6.1), which otherwise was not detected by NGS. Thus, given the well-documented efficacy of alectinib towards <i>EML4-ALK</i> v.1, combined second-line treatment with alectinib and the MET-TKI, crizotinib, was implemented resulting in very pronounced objective response, significantly improved quality of life, and no adverse events so far during the ongoing treatment (6 months).</p><p><strong>Conclusions: </strong>The combination of alectinib and crizotinib may be a feasible and effective treatment for <i>ALK</i>+ NSCLC with <i>de novo</i> <i>MET-</i>amplification. The latter may represent a mechanism of intrinsic ALK-TKI resistance and its recognition by FISH, in NGS-negative cases, may be considered before initiating first-line treatment. This recognition is clinically important as combined therapy with ALK-TKI and MET-inhibitor should be the preferred first-line treatment.</p>","PeriodicalId":23271,"journal":{"name":"Translational lung cancer research","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484720/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142475662","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}