4.7 Article

Multi-Omics Profiling Reveals Distinct Microenvironment Characterization and Suggests Immune Escape Mechanisms of Triple-Negative Breast Cancer

Journal

CLINICAL CANCER RESEARCH
Volume 25, Issue 16, Pages 5002-5014

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1078-0432.CCR-18-3524

Keywords

-

Categories

Funding

  1. National Natural Science Foundation of China [81874112, 81874113, 81572583, 81502278]
  2. Training Plan of Excellent Talents in Shanghai Municipality Health System [2017YQ038]
  3. Chen Guang project - Shanghai Municipal Education Commission [17CG01]
  4. Shanghai Education Development Foundation [17CG01]
  5. Shanghai Pujiang Program [18PJD007]
  6. Training Plan of Excellent Talents of Fudan University Shanghai Cancer Center [YJYQ201602]
  7. Municipal Project for Developing Emerging and Frontier Technology in Shanghai Hospitals [SHDC12010116]
  8. Cooperation Project of Conquering Major Diseases in Shanghai Municipality Health System [2013ZYJB0302]
  9. Innovation Team of Ministry of Education [IRT1223]
  10. Shanghai Key Laboratory of Breast Cancer [12DZ2260100]

Ask authors/readers for more resources

Purpose: The tumor microenvironment has a profound impact on prognosis and immunotherapy. However, the landscape of the triple-negative breast cancer (TNBC) microenvironment has not been fully understood. Experimental Design: Using the largest original multiomics dataset of TNBC (n = 386), we conducted an extensive immunogenomic analysis to explore the heterogeneity and prognostic significance of the TNBC microenvironment. We further analyzed the potential immune escape mechanisms of TNBC. Results: The TNBC microenvironment phenotypes were classified into three heterogeneous clusters: cluster 1, the immune-desert cluster, with low microenvironment cell infiltration; cluster 2, the innate immune-inactivated cluster, with resting innate immune cells and nonimmune stromal cells infiltration; and cluster 3, the immune-inflamed cluster, with abundant adaptive and innate immune cells infiltration. The clustering result was validated internally with pathologic sections and externally with The Cancer Genome Atlas and METABRIC cohorts. The microenvironment clusters had significant prognostic efficacy. In terms of potential immune escape mechanisms, cluster 1 was characterized by an incapability to attract immune cells, and MYC amplification was correlated with low immune infiltration. In cluster 2, chemotaxis but inactivation of innate immunity and low tumor antigen burden might contribute to immune escape, and mutations in the PI3K-AKT pathway might be correlated with this effect. Cluster 3 featured high expression of immune checkpoint molecules. Conclusions: Our study represents a step toward personalized immunotherapy for patients with TNBC. Immune checkpoint inhibitors might be effective for immune-inflamed cluster, and the transformation of cold tumors into hot tumors should be considered for immune-desert and innate immune-inactivated clusters.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available