Cargando…

Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer

Pleural effusion (PE) is commonly observed in advanced lung cancer and was suggested to contain both cell-free tumor DNA and tumor cells. Molecular profiling of PE represents a minimally invasive approach of detecting tumor driver mutations for clinical decision making, especially when tumor tissues...

Descripción completa

Detalles Bibliográficos
Autores principales: Tong, Lin, Ding, Ning, Tong, Xiaoling, Li, Jiamin, Zhang, Yong, Wang, Xiaodan, Xu, Xiaobo, Ye, Maosong, Li, Chun, Wu, Xue, Bao, Hairong, Zhang, Xin, Hong, Qunying, Song, Yuanlin, Shao, Yang W., Bai, Chunxue, Zhou, Jian, Hu, Jie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6735385/
https://www.ncbi.nlm.nih.gov/pubmed/31534501
http://dx.doi.org/10.7150/thno.34070
_version_ 1783450347309629440
author Tong, Lin
Ding, Ning
Tong, Xiaoling
Li, Jiamin
Zhang, Yong
Wang, Xiaodan
Xu, Xiaobo
Ye, Maosong
Li, Chun
Wu, Xue
Bao, Hairong
Zhang, Xin
Hong, Qunying
Song, Yuanlin
Shao, Yang W.
Bai, Chunxue
Zhou, Jian
Hu, Jie
author_facet Tong, Lin
Ding, Ning
Tong, Xiaoling
Li, Jiamin
Zhang, Yong
Wang, Xiaodan
Xu, Xiaobo
Ye, Maosong
Li, Chun
Wu, Xue
Bao, Hairong
Zhang, Xin
Hong, Qunying
Song, Yuanlin
Shao, Yang W.
Bai, Chunxue
Zhou, Jian
Hu, Jie
author_sort Tong, Lin
collection PubMed
description Pleural effusion (PE) is commonly observed in advanced lung cancer and was suggested to contain both cell-free tumor DNA and tumor cells. Molecular profiling of PE represents a minimally invasive approach of detecting tumor driver mutations for clinical decision making, especially when tumor tissues are not available. The objective of this study is to investigate the efficacy and precision of detecting gene alterations in PE samples to address the feasibility in clinical use. Methods: Sixty-three metastatic lung cancer patients with (n=30, cohort 1) or without (n=33, cohort 2) matched tumor tissues were enrolled in this study. PE and plasma samples of each patient were collected simultaneously. Supernatant and cell precipitate of PE were processed separately to extract cfDNA (PE-cfDNA) and sediment DNA (sDNA). All samples were subjected to targeted next-generation sequencing (NGS) of 416 cancer-related genes. Results: PE supernatants contain more abundant tumor DNA than PE sediments and plasma samples, suggested by higher mutant allele frequencies (MAF) and elevated mutation detection rate in PE-cfDNA (98.4% vs. 90.5% in PE sDNA vs. 87% in plasma cfDNA). In Cohort 1 with matched tumor tissue, tumor mutational burden (TMB) of PE-cfDNA was similar as tumor tissues (6.4 vs. 5.6), but significantly higher than PE sDNA (median TMB: 3.3) and plasma cfDNA (median TMB: 3.4). Ninety-three percent (27 out of 29) of tissue-determined driver mutations were detected in PE-cfDNA, including alterations in ALK, BRAF, EGFR, ERBB2, KRAS, NF1, PIK3CA, and RET, while only 62% were captured in plasma cfDNA. PE-cfDNA also has the highest detection rate of EGFR driver mutations in the full cohort (71% vs. 68% in PE sDNA vs. 59% in plasma cfDNA). Mutation detection from cytological negative and hemorrhagic PE is challenging. Comparatively, PE-cfDNA demonstrated absolute superiority than PE sDNA in such a scenario, suggesting that it is an independent source of tumor DNA and therefore less influenced by the abundance of tumor cells. Conclusion: Genomic profiling of PE-cfDNA offers an alternative, and potentially more meticulous approach in assessing tumor genomics in advanced lung cancer when tumor tissue is not available. Our data further demonstrate that in hemorrhagic or cytologically negative PE samples, PE-cfDNA has higher mutation detection sensitivity than sDNA and plasma cfDNA, and therefore is a more reliable source for genetic testing.
format Online
Article
Text
id pubmed-6735385
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-67353852019-09-18 Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer Tong, Lin Ding, Ning Tong, Xiaoling Li, Jiamin Zhang, Yong Wang, Xiaodan Xu, Xiaobo Ye, Maosong Li, Chun Wu, Xue Bao, Hairong Zhang, Xin Hong, Qunying Song, Yuanlin Shao, Yang W. Bai, Chunxue Zhou, Jian Hu, Jie Theranostics Research Paper Pleural effusion (PE) is commonly observed in advanced lung cancer and was suggested to contain both cell-free tumor DNA and tumor cells. Molecular profiling of PE represents a minimally invasive approach of detecting tumor driver mutations for clinical decision making, especially when tumor tissues are not available. The objective of this study is to investigate the efficacy and precision of detecting gene alterations in PE samples to address the feasibility in clinical use. Methods: Sixty-three metastatic lung cancer patients with (n=30, cohort 1) or without (n=33, cohort 2) matched tumor tissues were enrolled in this study. PE and plasma samples of each patient were collected simultaneously. Supernatant and cell precipitate of PE were processed separately to extract cfDNA (PE-cfDNA) and sediment DNA (sDNA). All samples were subjected to targeted next-generation sequencing (NGS) of 416 cancer-related genes. Results: PE supernatants contain more abundant tumor DNA than PE sediments and plasma samples, suggested by higher mutant allele frequencies (MAF) and elevated mutation detection rate in PE-cfDNA (98.4% vs. 90.5% in PE sDNA vs. 87% in plasma cfDNA). In Cohort 1 with matched tumor tissue, tumor mutational burden (TMB) of PE-cfDNA was similar as tumor tissues (6.4 vs. 5.6), but significantly higher than PE sDNA (median TMB: 3.3) and plasma cfDNA (median TMB: 3.4). Ninety-three percent (27 out of 29) of tissue-determined driver mutations were detected in PE-cfDNA, including alterations in ALK, BRAF, EGFR, ERBB2, KRAS, NF1, PIK3CA, and RET, while only 62% were captured in plasma cfDNA. PE-cfDNA also has the highest detection rate of EGFR driver mutations in the full cohort (71% vs. 68% in PE sDNA vs. 59% in plasma cfDNA). Mutation detection from cytological negative and hemorrhagic PE is challenging. Comparatively, PE-cfDNA demonstrated absolute superiority than PE sDNA in such a scenario, suggesting that it is an independent source of tumor DNA and therefore less influenced by the abundance of tumor cells. Conclusion: Genomic profiling of PE-cfDNA offers an alternative, and potentially more meticulous approach in assessing tumor genomics in advanced lung cancer when tumor tissue is not available. Our data further demonstrate that in hemorrhagic or cytologically negative PE samples, PE-cfDNA has higher mutation detection sensitivity than sDNA and plasma cfDNA, and therefore is a more reliable source for genetic testing. Ivyspring International Publisher 2019-07-28 /pmc/articles/PMC6735385/ /pubmed/31534501 http://dx.doi.org/10.7150/thno.34070 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Tong, Lin
Ding, Ning
Tong, Xiaoling
Li, Jiamin
Zhang, Yong
Wang, Xiaodan
Xu, Xiaobo
Ye, Maosong
Li, Chun
Wu, Xue
Bao, Hairong
Zhang, Xin
Hong, Qunying
Song, Yuanlin
Shao, Yang W.
Bai, Chunxue
Zhou, Jian
Hu, Jie
Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
title Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
title_full Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
title_fullStr Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
title_full_unstemmed Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
title_short Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
title_sort tumor-derived dna from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6735385/
https://www.ncbi.nlm.nih.gov/pubmed/31534501
http://dx.doi.org/10.7150/thno.34070
work_keys_str_mv AT tonglin tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT dingning tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT tongxiaoling tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT lijiamin tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT zhangyong tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT wangxiaodan tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT xuxiaobo tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT yemaosong tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT lichun tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT wuxue tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT baohairong tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT zhangxin tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT hongqunying tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT songyuanlin tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT shaoyangw tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT baichunxue tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT zhoujian tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer
AT hujie tumorderiveddnafrompleuraleffusionsupernatantasapromisingalternativetotumortissueingenomicprofilingofadvancedlungcancer