Cargando…

Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers

BACKGROUND: Immune checkpoint inhibitors (ICIs) have achieved impressive success in different cancer types, yet responses vary and predictive biomarkers are urgently needed. Growing evidence points to a link between DNA methylation and anti-tumor immunity, while clinical data on the association of g...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Hao-Xiang, Chen, Yan-Xing, Wang, Zi-Xian, Zhao, Qi, He, Ming-Ming, Wang, Ying-Nan, Wang, Feng, Xu, Rui-Hua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6798429/
https://www.ncbi.nlm.nih.gov/pubmed/31623662
http://dx.doi.org/10.1186/s40425-019-0737-3
_version_ 1783460038887604224
author Wu, Hao-Xiang
Chen, Yan-Xing
Wang, Zi-Xian
Zhao, Qi
He, Ming-Ming
Wang, Ying-Nan
Wang, Feng
Xu, Rui-Hua
author_facet Wu, Hao-Xiang
Chen, Yan-Xing
Wang, Zi-Xian
Zhao, Qi
He, Ming-Ming
Wang, Ying-Nan
Wang, Feng
Xu, Rui-Hua
author_sort Wu, Hao-Xiang
collection PubMed
description BACKGROUND: Immune checkpoint inhibitors (ICIs) have achieved impressive success in different cancer types, yet responses vary and predictive biomarkers are urgently needed. Growing evidence points to a link between DNA methylation and anti-tumor immunity, while clinical data on the association of genomic alterations in DNA methylation-related genes and ICI response are lacking. METHODS: Clinical cohorts with annotated response and survival data and matched mutational data from published studies were collected and consolidated. The predictive function of specific mutated genes was first tested in the discovery cohort and later validated in the validation cohort. The association between specific mutated genes and tumor immunogenicity and anti-tumor immunity was further investigated in the Cancer Genome Altas (TCGA) dataset. RESULTS: Among twenty-one key genes involving in the regulation of DNA methylation, TET1-mutant (TET1-MUT) was enriched in patients responding to ICI treatment in the discovery cohort (P < 0.001). TET1 was recurrently mutated across multiple cancers and more frequently seen in skin, lung, gastrointestinal, and urogenital cancers. In the discovery cohort (n = 519), significant differences were observed between TET1-MUT and TET1-wildtype (TET1-WT) patients regarding objective response rate (ORR, 60.9% versus 22.8%, P < 0.001), durable clinical benefit (DCB, 71.4% versus 31.6%, P < 0.001), and progression-free survival (PFS, hazard ratio = 0.46 [95% confidence interval, 0.25 to 0.82], P = 0.008). In the validation cohort (n = 1395), significant overall survival (OS) benefit was detected in the TET1-MUT patients compared to TET1-WT patients (hazard ratio = 0.47 [95% confidence interval, 0.25 to 0.88], P = 0.019), which was, importantly, independent of tumor mutational burden and high microsatellite instability; as well as not attributed to the prognostic impact of TET1-MUT (P > 0.05 in both two non-ICI-treated cohorts). In TCGA dataset, TET1-MUT was strongly associated with higher tumor mutational burden and neoantigen load, and inflamed pattern of tumor-infiltrating T lymphocytes, immune signatures and immune-related gene expressions. CONCLUSIONS: TET1-MUT was strongly associated with higher ORR, better DCB, longer PFS, and improved OS in patients receiving ICI treatment, suggesting that TET1-MUT is a novel predictive biomarker for immune checkpoint blockade across multiple cancer types.
format Online
Article
Text
id pubmed-6798429
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-67984292019-10-21 Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers Wu, Hao-Xiang Chen, Yan-Xing Wang, Zi-Xian Zhao, Qi He, Ming-Ming Wang, Ying-Nan Wang, Feng Xu, Rui-Hua J Immunother Cancer Research Article BACKGROUND: Immune checkpoint inhibitors (ICIs) have achieved impressive success in different cancer types, yet responses vary and predictive biomarkers are urgently needed. Growing evidence points to a link between DNA methylation and anti-tumor immunity, while clinical data on the association of genomic alterations in DNA methylation-related genes and ICI response are lacking. METHODS: Clinical cohorts with annotated response and survival data and matched mutational data from published studies were collected and consolidated. The predictive function of specific mutated genes was first tested in the discovery cohort and later validated in the validation cohort. The association between specific mutated genes and tumor immunogenicity and anti-tumor immunity was further investigated in the Cancer Genome Altas (TCGA) dataset. RESULTS: Among twenty-one key genes involving in the regulation of DNA methylation, TET1-mutant (TET1-MUT) was enriched in patients responding to ICI treatment in the discovery cohort (P < 0.001). TET1 was recurrently mutated across multiple cancers and more frequently seen in skin, lung, gastrointestinal, and urogenital cancers. In the discovery cohort (n = 519), significant differences were observed between TET1-MUT and TET1-wildtype (TET1-WT) patients regarding objective response rate (ORR, 60.9% versus 22.8%, P < 0.001), durable clinical benefit (DCB, 71.4% versus 31.6%, P < 0.001), and progression-free survival (PFS, hazard ratio = 0.46 [95% confidence interval, 0.25 to 0.82], P = 0.008). In the validation cohort (n = 1395), significant overall survival (OS) benefit was detected in the TET1-MUT patients compared to TET1-WT patients (hazard ratio = 0.47 [95% confidence interval, 0.25 to 0.88], P = 0.019), which was, importantly, independent of tumor mutational burden and high microsatellite instability; as well as not attributed to the prognostic impact of TET1-MUT (P > 0.05 in both two non-ICI-treated cohorts). In TCGA dataset, TET1-MUT was strongly associated with higher tumor mutational burden and neoantigen load, and inflamed pattern of tumor-infiltrating T lymphocytes, immune signatures and immune-related gene expressions. CONCLUSIONS: TET1-MUT was strongly associated with higher ORR, better DCB, longer PFS, and improved OS in patients receiving ICI treatment, suggesting that TET1-MUT is a novel predictive biomarker for immune checkpoint blockade across multiple cancer types. BioMed Central 2019-10-17 /pmc/articles/PMC6798429/ /pubmed/31623662 http://dx.doi.org/10.1186/s40425-019-0737-3 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Wu, Hao-Xiang
Chen, Yan-Xing
Wang, Zi-Xian
Zhao, Qi
He, Ming-Ming
Wang, Ying-Nan
Wang, Feng
Xu, Rui-Hua
Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers
title Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers
title_full Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers
title_fullStr Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers
title_full_unstemmed Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers
title_short Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers
title_sort alteration in tet1 as potential biomarker for immune checkpoint blockade in multiple cancers
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6798429/
https://www.ncbi.nlm.nih.gov/pubmed/31623662
http://dx.doi.org/10.1186/s40425-019-0737-3
work_keys_str_mv AT wuhaoxiang alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT chenyanxing alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT wangzixian alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT zhaoqi alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT hemingming alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT wangyingnan alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT wangfeng alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers
AT xuruihua alterationintet1aspotentialbiomarkerforimmunecheckpointblockadeinmultiplecancers