Comparative analysis of microsatellite instability-high (MSI-H) BRAF V600E-mutated versus MSI-H BRAFwild type colorectal cancers (CRC), including tumor microenvironment (TME), associated genomic alterations, and immunometabolomic biomarkers.

Authors

Mohamed Salem

Mohamed E. Salem

Levine Cancer Institute, Atrium Health, Charlotte, NC

Mohamed E. Salem , Scott Kopetz , Sherif Mohamed El-Refai , Josep Tabernero , Frank A. Sinicrope , Jeanne Tie , Thomas J. George , Eric VanCutsem , Elizabeth Mauer , Sara Lonardi , Thierry Andre , Michael J. Overman , David Foureau

Organizations

Levine Cancer Institute, Atrium Health, Charlotte, NC, The University of Texas MD Anderson Cancer Center, Houston, TX, Tempus Labs, Chicago, IL, Vall d’Hebron Institute of Oncology, Barcelona, Spain, Mayo Clinic, Rochester, MN, Peter MacCallum Cancer Centre, University of Melbourne, Walter and Eliza Hall Institute, Melbourne, VIC, Australia, The University of Florida Health Cancer Center, Gainesville, FL, University of Leuven, Leuven, Belgium, Veneto Institute of Oncology, IRCCS, Padua, Italy, Sorbonne University, Saint-Antoine Hospital, AP-HP, Paris, France, University of Texas MD Anderson Cancer Center and SWOG, Houston, TX

Research Funding

No funding received

Background: The BRAFV600E mutation is associated with the hypermethylator phenotype CIMP, which can also lead to the MSI-H phenotype. BRAFV600E mutation and MSI-H/dMMR status seem to be biologically intertwined; however, the impact of coexisting BRAFV600E mutations on the TME and immunometabolomic features of MSI-H/dMMR CRC tumors is not well characterized. Methods: A retrospective review of deidentified records of patients with MSI-H/dMMR CRC tumors was conducted using next-generation sequencing data (Tempus |xT assay: DNA-seq of 595-648 genes at 500x coverage, and full transcriptome RNA-seq). Several immune markers of tumor immunogenicity in BRAF wild-type (BRAFwt) vs. V600E-mutated (BRAFV600E) tumors were assessed, including tumor mutational burden (TMB), neoantigen tumor burden (NTB, ScanNeo), PD-L1 expression, immune infiltration, and canonical immuno-metabolomic pathways (82 geneset signatures). Results: A total of 459 MSI-H/dMMR CRC tumors were analyzed, of which 123 (27%) tumors harbored BRAFV600Emutations, and 336 (73%) were BRAFwt. MSI-H/dMMR BRAFV600E tumors were more frequently identified in females (69% vs. 55%; P= 0.01), non-Hispanic or non-Latino (100% vs. 73%; P =0.001), and older patients (median age: 76 yrs vs. 62 yrs; P< 0.001). Compared to BRAFWT, BRAFV600E tumors exhibited significantly higher TMB (Median: 49 mut/MB vs. 36 mut/MB; P< 0.001) and were more frequently associated with TMB-High status (> 10 mut/MB; 100% vs. 95%; P = 0.008); however, no significant differences were observed with tumor NTB, immune score, or T cell infiltration (CD4 or CD8). NK cell infiltration was higher in the BRAFV600Ecohort (< 0.001). When compared to BRAFWT tumors, BRAFV600E tumors harbored a greater frequency of mutations in MSH6(42% vs. 20%), B2M (33% vs. 16%), BRCA2 (31% vs. 12%), ATM (23% vs. 12%), and TP53 (30% vs. 19%) but a lower frequency of MSH2 (3.3% vs. 11%), all P< 0.05. Pathway enrichment analysis identified 10 significantly altered signaling pathways, most of which related to stromal/immune cell signaling and metabolism. Five were upregulated among BRAFV600E tumors: glycerophospholipid, galactose, cyclin-dependent cell signaling; Nucleotide, and TH1 inflammation. Five pathways were downregulated (Wnt, Notch, TH17 inflammation, amino sugar, and cancer stem cell signaling). Conclusions: MSI-H/dMMR BRAFV600E CRCs undergo broad metabolic reprogramming (e.g., glycerophospholipidgalactose, nucleotide). A rise in lipid metabolism, particularly with NK inflammation, suggests that BRAFV600Emutated tumors may be associated with a non-classical immune component. BRAFV600E and BRAFwt CRCs exhibited similar NTB and T cell infiltration, suggesting that both are likely to benefit from immune checkpoint inhibitors.

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2022 ASCO Annual Meeting

Session Type

Poster Session

Session Title

Developmental Therapeutics—Molecularly Targeted Agents and Tumor Biology

Track

Developmental Therapeutics—Molecularly Targeted Agents and Tumor Biology

Sub Track

Molecular Diagnostics and Imaging

Citation

J Clin Oncol 40, 2022 (suppl 16; abstr 3066)

DOI

10.1200/JCO.2022.40.16_suppl.3066

Abstract #

3066

Poster Bd #

58

Abstract Disclosures

Similar Abstracts

First Author: Jeanne Tie

First Author: Parvathi Myer

Abstract

2024 ASCO Gastrointestinal Cancers Symposium

Incidence and genomic characteristics of gene fusions in a large Chinese colorectal cancer cohort.

First Author: Furong Kou