Cargando…

Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer

BACKGROUND: Autophagy is a highly conserved cellular proteolytic process that can interact with innate immune signaling pathways to affect the growth of tumor cells. However, the regulatory mechanism of autophagy in the tumor microenvironment, drug sensitivity, and immunotherapy is still unclear. ME...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Yuwei, Deng, Zhibo, Sun, Yang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9890868/
https://www.ncbi.nlm.nih.gov/pubmed/36721247
http://dx.doi.org/10.1186/s41065-023-00263-2
_version_ 1784881027891593216
author Chen, Yuwei
Deng, Zhibo
Sun, Yang
author_facet Chen, Yuwei
Deng, Zhibo
Sun, Yang
author_sort Chen, Yuwei
collection PubMed
description BACKGROUND: Autophagy is a highly conserved cellular proteolytic process that can interact with innate immune signaling pathways to affect the growth of tumor cells. However, the regulatory mechanism of autophagy in the tumor microenvironment, drug sensitivity, and immunotherapy is still unclear. METHODS: Based on the prognostic autophagy-related genes, we used the unsupervised clustering method to divide 866 ovarian cancer samples into two regulatory patterns. According to the phenotypic regulation pattern formed by the differential gene between the two regulation patterns, a risk model was constructed to quantify patients with ovarian cancer. Then, we systematically analyzed the relationship between the risk model and immune cell infiltration, immunotherapeutic response, and drug sensitivity. RESULTS: Based on autophagy-related genes, we found two autophagy regulation patterns, and confirmed that there were differences in prognosis and immune cell infiltration between them. Subsequently, we constructed a risk model, which was divided into a high-risk group and a low-risk group. We found that the high-risk group had a worse prognosis, and the main infiltrating immune cells were adaptive immune cells, such as Th2 cells, Tgd cells, eosinophils cells, and lymph vessels cells. The low-risk group had a better prognosis, and the most infiltrated immune cells were innate immune cells, such as aDC cells, NK CD56dim cells, and NK CD56bright cells. Furthermore, we found that the risk model could predict chemosensitivity and immunotherapy response, suggesting that the risk model may help to formulate personalized treatment plans for patients. CONCLUSIONS: Our study comprehensively analyzed the prognostic potential of autophagy-related risk models in ovarian cancer and determined their clinical guiding role in targeted therapy and immunotherapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s41065-023-00263-2.
format Online
Article
Text
id pubmed-9890868
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-98908682023-02-02 Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer Chen, Yuwei Deng, Zhibo Sun, Yang Hereditas Research BACKGROUND: Autophagy is a highly conserved cellular proteolytic process that can interact with innate immune signaling pathways to affect the growth of tumor cells. However, the regulatory mechanism of autophagy in the tumor microenvironment, drug sensitivity, and immunotherapy is still unclear. METHODS: Based on the prognostic autophagy-related genes, we used the unsupervised clustering method to divide 866 ovarian cancer samples into two regulatory patterns. According to the phenotypic regulation pattern formed by the differential gene between the two regulation patterns, a risk model was constructed to quantify patients with ovarian cancer. Then, we systematically analyzed the relationship between the risk model and immune cell infiltration, immunotherapeutic response, and drug sensitivity. RESULTS: Based on autophagy-related genes, we found two autophagy regulation patterns, and confirmed that there were differences in prognosis and immune cell infiltration between them. Subsequently, we constructed a risk model, which was divided into a high-risk group and a low-risk group. We found that the high-risk group had a worse prognosis, and the main infiltrating immune cells were adaptive immune cells, such as Th2 cells, Tgd cells, eosinophils cells, and lymph vessels cells. The low-risk group had a better prognosis, and the most infiltrated immune cells were innate immune cells, such as aDC cells, NK CD56dim cells, and NK CD56bright cells. Furthermore, we found that the risk model could predict chemosensitivity and immunotherapy response, suggesting that the risk model may help to formulate personalized treatment plans for patients. CONCLUSIONS: Our study comprehensively analyzed the prognostic potential of autophagy-related risk models in ovarian cancer and determined their clinical guiding role in targeted therapy and immunotherapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s41065-023-00263-2. BioMed Central 2023-02-01 /pmc/articles/PMC9890868/ /pubmed/36721247 http://dx.doi.org/10.1186/s41065-023-00263-2 Text en © The Author(s) 2023 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
Chen, Yuwei
Deng, Zhibo
Sun, Yang
Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
title Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
title_full Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
title_fullStr Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
title_full_unstemmed Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
title_short Development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
title_sort development of a risk model based on autophagy-related genes to predict survival and immunotherapy response in ovarian cancer
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9890868/
https://www.ncbi.nlm.nih.gov/pubmed/36721247
http://dx.doi.org/10.1186/s41065-023-00263-2
work_keys_str_mv AT chenyuwei developmentofariskmodelbasedonautophagyrelatedgenestopredictsurvivalandimmunotherapyresponseinovariancancer
AT dengzhibo developmentofariskmodelbasedonautophagyrelatedgenestopredictsurvivalandimmunotherapyresponseinovariancancer
AT sunyang developmentofariskmodelbasedonautophagyrelatedgenestopredictsurvivalandimmunotherapyresponseinovariancancer