Cargando…

Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer

BACKGROUND: Immunotherapy has emerged as a new cancer treatment modality. However, tumour heterogeneity can diminish checkpoint blockade response and shorten patient survival. As a source of tumour heterogeneity, cancer stem cells act as an indispensable reservoir for local recurrence and distant me...

Descripción completa

Detalles Bibliográficos
Autores principales: Shi, Haojun, Tsang, Yiusing, Yang, Yisi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9733357/
https://www.ncbi.nlm.nih.gov/pubmed/36494785
http://dx.doi.org/10.1186/s12885-022-10397-7
_version_ 1784846356079181824
author Shi, Haojun
Tsang, Yiusing
Yang, Yisi
author_facet Shi, Haojun
Tsang, Yiusing
Yang, Yisi
author_sort Shi, Haojun
collection PubMed
description BACKGROUND: Immunotherapy has emerged as a new cancer treatment modality. However, tumour heterogeneity can diminish checkpoint blockade response and shorten patient survival. As a source of tumour heterogeneity, cancer stem cells act as an indispensable reservoir for local recurrence and distant metastasis. Thus, precision immunotherapy targeting tumour heterogeneity requires a comprehensive understanding of cancer stem cell immunology. Our study aimed to identify stemness-related inhibitory immune checkpoints and relevant regulatory pathways in pancreatic cancer. METHODS: Pancreatic cancer-specific datasets in The Cancer Genome Atlas and the Cancer Therapeutics Response Portal were collected for in-depth bioinformatic analysis. Differentially expressed genes between pancreatic cancers with high and low stemness index (mRNAsi) scores were compared to screen out inhibitory immune checkpoints. Survival analysis was used to predict the prognostic value of immune checkpoint plus immune infiltrate in patients with pancreatic cancer. The expression of stemness-related immune checkpoint across immune subtypes of pancreatic cancer was detected and gene set enrichment analysis was performed to figure out the relevant regulatory signallings. RESULTS: The abundance of cancer stemness predicted a low immunotherapy response to pancreatic cancer. The inhibitory immune checkpoint CEACAM5 that was enriched in pancreatic cancers with high mRNAsi scores also exhibited a strong correlation with invasive cell-enriched signature and Msi(+) tumour-initiating cell-enriched signature. Levels of CEACAM5 expression were higher in the interferon-γ dominant immune subtype of pancreatic cancers that are characterized by high M1 macrophage infiltration. The patient group with high levels of CEACAM5 expression had a high risk of poor overall survival, even if accompanied by high infiltration of M1 macrophages. Furthermore, prostanoid and long-chain unsaturated fatty acid metabolic processes showed a significant association with cancer stemness and CEACAM5 expression. CONCLUSIONS: Our findings suggest that CEACAM5 is a candidate stemness-related innate immune checkpoint in pancreatic cancer, and is potentially regulated by prostanoid and long-chain unsaturated fatty acid metabolic processes. Immune checkpoint blockade of CEACAM5, which synergizes with inhibition of those regulatory pathways, may improve the efficacy of precision immunotherapy targeting tumour heterogeneity caused by cancer stem cells. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12885-022-10397-7.
format Online
Article
Text
id pubmed-9733357
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-97333572022-12-10 Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer Shi, Haojun Tsang, Yiusing Yang, Yisi BMC Cancer Research BACKGROUND: Immunotherapy has emerged as a new cancer treatment modality. However, tumour heterogeneity can diminish checkpoint blockade response and shorten patient survival. As a source of tumour heterogeneity, cancer stem cells act as an indispensable reservoir for local recurrence and distant metastasis. Thus, precision immunotherapy targeting tumour heterogeneity requires a comprehensive understanding of cancer stem cell immunology. Our study aimed to identify stemness-related inhibitory immune checkpoints and relevant regulatory pathways in pancreatic cancer. METHODS: Pancreatic cancer-specific datasets in The Cancer Genome Atlas and the Cancer Therapeutics Response Portal were collected for in-depth bioinformatic analysis. Differentially expressed genes between pancreatic cancers with high and low stemness index (mRNAsi) scores were compared to screen out inhibitory immune checkpoints. Survival analysis was used to predict the prognostic value of immune checkpoint plus immune infiltrate in patients with pancreatic cancer. The expression of stemness-related immune checkpoint across immune subtypes of pancreatic cancer was detected and gene set enrichment analysis was performed to figure out the relevant regulatory signallings. RESULTS: The abundance of cancer stemness predicted a low immunotherapy response to pancreatic cancer. The inhibitory immune checkpoint CEACAM5 that was enriched in pancreatic cancers with high mRNAsi scores also exhibited a strong correlation with invasive cell-enriched signature and Msi(+) tumour-initiating cell-enriched signature. Levels of CEACAM5 expression were higher in the interferon-γ dominant immune subtype of pancreatic cancers that are characterized by high M1 macrophage infiltration. The patient group with high levels of CEACAM5 expression had a high risk of poor overall survival, even if accompanied by high infiltration of M1 macrophages. Furthermore, prostanoid and long-chain unsaturated fatty acid metabolic processes showed a significant association with cancer stemness and CEACAM5 expression. CONCLUSIONS: Our findings suggest that CEACAM5 is a candidate stemness-related innate immune checkpoint in pancreatic cancer, and is potentially regulated by prostanoid and long-chain unsaturated fatty acid metabolic processes. Immune checkpoint blockade of CEACAM5, which synergizes with inhibition of those regulatory pathways, may improve the efficacy of precision immunotherapy targeting tumour heterogeneity caused by cancer stem cells. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12885-022-10397-7. BioMed Central 2022-12-09 /pmc/articles/PMC9733357/ /pubmed/36494785 http://dx.doi.org/10.1186/s12885-022-10397-7 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Shi, Haojun
Tsang, Yiusing
Yang, Yisi
Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
title Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
title_full Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
title_fullStr Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
title_full_unstemmed Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
title_short Identification of CEACAM5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
title_sort identification of ceacam5 as a stemness-related inhibitory immune checkpoint in pancreatic cancer
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9733357/
https://www.ncbi.nlm.nih.gov/pubmed/36494785
http://dx.doi.org/10.1186/s12885-022-10397-7
work_keys_str_mv AT shihaojun identificationofceacam5asastemnessrelatedinhibitoryimmunecheckpointinpancreaticcancer
AT tsangyiusing identificationofceacam5asastemnessrelatedinhibitoryimmunecheckpointinpancreaticcancer
AT yangyisi identificationofceacam5asastemnessrelatedinhibitoryimmunecheckpointinpancreaticcancer