Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/197332
Title: A novel gene signature unveils three distinct immune- metabolic rewiring patterns conserved across diverse tumor types and associated with outcomes
Author: Pedrosa, Leire
Foguet Coll, Carles
Oliveres, Helena
Archilla, Iván
García de Herreros, Marta
Rodríguez, Adela
Postigo, Antonio
Benítez-Ribas, Daniel
Camps, Jordi
Cuatrecasas Freixas, Miriam
Castells Garangou, Antoni
Prat Aparicio, Aleix
Thomson, Timothy M.
Maurel Santasusana, Joan
Cascante i Serratosa, Marta
Keywords: Càncer colorectal
Metàstasi
Marcadors bioquímics
Immunoteràpia
Metabolisme
Colorectal cancer
Metastasis
Biochemical markers
Immunotheraphy
Metabolism
Issue Date: 2-Sep-2022
Publisher: Frontiers Media
Abstract: Existing immune signatures and tumor mutational burden have only modest predictive capacity for the efficacy of immune check point inhibitors. In this study, we developed an immune-metabolic signature suitable for personalized ICI therapies. A classifier using an immune-metabolic signature (IMMETCOLS) was developed on a training set of 77 metastatic colorectal cancer (mCRC) samples and validated on 4,200 tumors from the TCGA database belonging to 11 types. Here, we reveal that the IMMETCOLS signature classifies tumors into three distinct immune-metabolic clusters. Cluster 1 displays markers of enhanced glycolisis, hexosamine byosinthesis and epithelial-to-mesenchymal transition. On multivariate analysis, cluster 1 tumors were enriched in pro-immune signature but not in immunophenoscore and were associated with the poorest median survival. Its predicted tumor metabolic features suggest an acidic-lactate-rich tumor microenvironment (TME) geared to an immunosuppressive setting, enriched in fibroblasts. Cluster 2 displays features of gluconeogenesis ability, which is needed for glucose-independent survival and preferential use of alternative carbon sources, including glutamine and lipid uptake/β-oxidation. Its metabolic features suggest a hypoxic and hypoglycemic TME, associated with poor tumor-associated antigen presentation. Finally, cluster 3 is highly glycolytic but also has a solid mitochondrial function, with concomitant upregulation of glutamine and essential amino acid transporters and the pentose phosphate pathway leading to glucose exhaustion in the TME and immunosuppression. Together, these findings suggest that the IMMETCOLS signature provides a classifier of tumors from diverse origins, yielding three clusters with distinct immune-metabolic profiles, representing a new predictive tool for patient selection for specific immune-metabolic therapeutic approaches.
Note: Reproducció del document publicat a: https://doi.org/10.3389/fimmu.2022.926304
It is part of: Frontiers in Immunology, 2022, vol. 13, p. 926304
URI: http://hdl.handle.net/2445/197332
Related resource: https://doi.org/10.3389/fimmu.2022.926304
ISSN: 1664-3224
Appears in Collections:Articles publicats en revistes (IDIBAPS: Institut d'investigacions Biomèdiques August Pi i Sunyer)
Articles publicats en revistes (Fonaments Clínics)

Files in This Item:
File Description SizeFormat 
729686.pdf17.76 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons