Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer

BackgroundData supporting high tumor mutational burden (TMB-H) as a lone biomarker for an immune-responsive tumor microenvironment (TME) in metastatic breast cancer (MBC) are weak, yet tumor agnostic approval in TMB-H advanced tumors provides immune checkpoint inhibition (ICI) as a clinical option....

Full description

Bibliographic Details
Main Authors: Sarah Sammons, Andrew Elliott, Romualdo Barroso-Sousa, Saranya Chumsri, Antoinette R. Tan, George W. Sledge, Sara M. Tolaney, Evanthia T. Roussos Torres
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-08-01
Series:Frontiers in Oncology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fonc.2023.1235902/full
_version_ 1797744482072920064
author Sarah Sammons
Sarah Sammons
Sarah Sammons
Andrew Elliott
Romualdo Barroso-Sousa
Romualdo Barroso-Sousa
Saranya Chumsri
Antoinette R. Tan
George W. Sledge
Sara M. Tolaney
Sara M. Tolaney
Sara M. Tolaney
Evanthia T. Roussos Torres
author_facet Sarah Sammons
Sarah Sammons
Sarah Sammons
Andrew Elliott
Romualdo Barroso-Sousa
Romualdo Barroso-Sousa
Saranya Chumsri
Antoinette R. Tan
George W. Sledge
Sara M. Tolaney
Sara M. Tolaney
Sara M. Tolaney
Evanthia T. Roussos Torres
author_sort Sarah Sammons
collection DOAJ
description BackgroundData supporting high tumor mutational burden (TMB-H) as a lone biomarker for an immune-responsive tumor microenvironment (TME) in metastatic breast cancer (MBC) are weak, yet tumor agnostic approval in TMB-H advanced tumors provides immune checkpoint inhibition (ICI) as a clinical option. We evaluated concurrent predictors of immune-responsive and non-responsive TME within MBC.MethodsTumor samples from patients with MBC (N=5621) were analyzed by next-generation sequencing of DNA (592-gene panel or whole exome) and RNA (whole transcriptome) at Caris Life Sciences (Phoenix, AZ). TMB-H threshold was set to ≥ 10 muts/Mb. PDL-1 was evaluated using SP142 antibody. Gene expression profiling and RNA deconvolution were used to estimate immune and stromal cell population abundance in the TME, and transcriptomic signature of immunotherapy response (T cell-inflamed score).Results461 (8.2%) TMB-H MBC samples were identified. Consistent with prior studies, TMB-H tumors exhibited significant dMMR/MSI-H enrichment (7 vs. 0%, p<0.0001) and PD-L1+ expression (36 vs. 28%, p<0.05) compared to TMB-L. Across all samples, T cell-inflamed scores were weakly correlated with TMB. TMB-H was not associated with significantly increased immune responsive cell types (CD8+ T-cells, NK cells, or B cells) or immune response gene signatures (e.g. antigen presentation), yet positive trends were observed, while immunosuppressive fibroblasts were significantly decreased in TMB-H tumors (0.84-fold change compared to TMB-L, P<0.05). HR+/HER2- breast cancer was the only subtype in which TMB-H tumors exhibited increased T cell-inflamed scores vs. TMB-L. Concurrent PD-L1+ or dMMR/MSI-H with TMB-H was associated with high T cell-inflamed scores in both HR+/HER2- and TNBC. Among several associated biomarkers, B2M mutations and CD274 amplifications were positively associated with T-cell inflamed scores in TMB-H tumors; CDH1 and ERBB2 mutations were negatively associated.ConclusionHigh TMB alone does not strongly correlate with immune infiltrate or immune-related gene signatures in MBC. TMB-H predicts T-cell inflamed signature compared to TMB-L in HR+/HER2- tumors only. Along with MSI-H and PD-L1+, several biomarkers, including B2M mutation and CD274 amplification, may help predict ICI benefit amongst TMB-H tumors. Co-occurring biomarkers within TMB-H breast cancer warrant evaluation in larger cohorts for response or resistance to ICI to develop composite predictive biomarkers in MBC.
first_indexed 2024-03-12T15:10:18Z
format Article
id doaj.art-8b0d35ab3ce5400f8a02d8f094918d24
institution Directory Open Access Journal
issn 2234-943X
language English
last_indexed 2024-03-12T15:10:18Z
publishDate 2023-08-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Oncology
spelling doaj.art-8b0d35ab3ce5400f8a02d8f094918d242023-08-11T17:53:30ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2023-08-011310.3389/fonc.2023.12359021235902Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancerSarah Sammons0Sarah Sammons1Sarah Sammons2Andrew Elliott3Romualdo Barroso-Sousa4Romualdo Barroso-Sousa5Saranya Chumsri6Antoinette R. Tan7George W. Sledge8Sara M. Tolaney9Sara M. Tolaney10Sara M. Tolaney11Evanthia T. Roussos Torres12Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United StatesBreast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, United StatesHarvard Medical School, Boston, MA, United StatesClinical and Translational Research, Caris Life Sciences, Phoenix, AZ, United StatesDepartment of Oncology, Dasa Institute for Education and Research (IEPD), Brasilia, BrazilDasa Oncology/Hospital Brasilia, Brasilia, BrazilDepartment of Hematology Oncology and Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United StatesLevine Cancer Institute, Atrium Health, Charlotte, NC, United StatesClinical and Translational Research, Caris Life Sciences, Phoenix, AZ, United StatesDepartment of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United StatesBreast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, United StatesHarvard Medical School, Boston, MA, United StatesDivision of Oncology, Department of Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United StatesBackgroundData supporting high tumor mutational burden (TMB-H) as a lone biomarker for an immune-responsive tumor microenvironment (TME) in metastatic breast cancer (MBC) are weak, yet tumor agnostic approval in TMB-H advanced tumors provides immune checkpoint inhibition (ICI) as a clinical option. We evaluated concurrent predictors of immune-responsive and non-responsive TME within MBC.MethodsTumor samples from patients with MBC (N=5621) were analyzed by next-generation sequencing of DNA (592-gene panel or whole exome) and RNA (whole transcriptome) at Caris Life Sciences (Phoenix, AZ). TMB-H threshold was set to ≥ 10 muts/Mb. PDL-1 was evaluated using SP142 antibody. Gene expression profiling and RNA deconvolution were used to estimate immune and stromal cell population abundance in the TME, and transcriptomic signature of immunotherapy response (T cell-inflamed score).Results461 (8.2%) TMB-H MBC samples were identified. Consistent with prior studies, TMB-H tumors exhibited significant dMMR/MSI-H enrichment (7 vs. 0%, p<0.0001) and PD-L1+ expression (36 vs. 28%, p<0.05) compared to TMB-L. Across all samples, T cell-inflamed scores were weakly correlated with TMB. TMB-H was not associated with significantly increased immune responsive cell types (CD8+ T-cells, NK cells, or B cells) or immune response gene signatures (e.g. antigen presentation), yet positive trends were observed, while immunosuppressive fibroblasts were significantly decreased in TMB-H tumors (0.84-fold change compared to TMB-L, P<0.05). HR+/HER2- breast cancer was the only subtype in which TMB-H tumors exhibited increased T cell-inflamed scores vs. TMB-L. Concurrent PD-L1+ or dMMR/MSI-H with TMB-H was associated with high T cell-inflamed scores in both HR+/HER2- and TNBC. Among several associated biomarkers, B2M mutations and CD274 amplifications were positively associated with T-cell inflamed scores in TMB-H tumors; CDH1 and ERBB2 mutations were negatively associated.ConclusionHigh TMB alone does not strongly correlate with immune infiltrate or immune-related gene signatures in MBC. TMB-H predicts T-cell inflamed signature compared to TMB-L in HR+/HER2- tumors only. Along with MSI-H and PD-L1+, several biomarkers, including B2M mutation and CD274 amplification, may help predict ICI benefit amongst TMB-H tumors. Co-occurring biomarkers within TMB-H breast cancer warrant evaluation in larger cohorts for response or resistance to ICI to develop composite predictive biomarkers in MBC.https://www.frontiersin.org/articles/10.3389/fonc.2023.1235902/fullbreast cancertumor mutational burdengenetic profilingmicroenvironmentimmune checkpoint inhibitors
spellingShingle Sarah Sammons
Sarah Sammons
Sarah Sammons
Andrew Elliott
Romualdo Barroso-Sousa
Romualdo Barroso-Sousa
Saranya Chumsri
Antoinette R. Tan
George W. Sledge
Sara M. Tolaney
Sara M. Tolaney
Sara M. Tolaney
Evanthia T. Roussos Torres
Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer
Frontiers in Oncology
breast cancer
tumor mutational burden
genetic profiling
microenvironment
immune checkpoint inhibitors
title Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer
title_full Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer
title_fullStr Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer
title_full_unstemmed Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer
title_short Concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden-high breast cancer
title_sort concurrent predictors of an immune responsive tumor microenvironment within tumor mutational burden high breast cancer
topic breast cancer
tumor mutational burden
genetic profiling
microenvironment
immune checkpoint inhibitors
url https://www.frontiersin.org/articles/10.3389/fonc.2023.1235902/full
work_keys_str_mv AT sarahsammons concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT sarahsammons concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT sarahsammons concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT andrewelliott concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT romualdobarrososousa concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT romualdobarrososousa concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT saranyachumsri concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT antoinettertan concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT georgewsledge concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT saramtolaney concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT saramtolaney concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT saramtolaney concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer
AT evanthiatroussostorres concurrentpredictorsofanimmuneresponsivetumormicroenvironmentwithintumormutationalburdenhighbreastcancer