4.8 Article

EGFR is a master switch between immunosuppressive and immunoactive tumor microenvironment in inflammatory breast cancer

Journal

SCIENCE ADVANCES
Volume 8, Issue 50, Pages -

Publisher

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/sciadv.abn7983

Keywords

-

Funding

  1. Breast Cancer Research Foundation [BCRF-17-161]
  2. NIH [1R01CA20504301A1, 1R01CA258523-01A1]
  3. Nylene Eckles Distinguished Professor in Breast Cancer Research
  4. Morgan Welch Inflammatory Breast Cancer Research Program and Clinic
  5. State of Texas Rare and Aggressive Breast Cancer Research Program
  6. IBC Network Foundation
  7. Emerson Collective Cancer Research Fund [689199]
  8. Cathy R Smith Immunotherapy Acceleration Award
  9. Grand Prix Ruban Rose
  10. Label Ligue Cancer du Sein
  11. NIH through MD Anderson's Cancer Center Support Grant [CA016672]
  12. NCI Research Specialist Award [1R50 CA243707-01A1]
  13. CPRIT SINGLE CORE Facilities [RP180684]

Ask authors/readers for more resources

This study reveals that EGFR-targeted therapy can remodel the tumor microenvironment of inflammatory breast cancer, leading to increased cytotoxic T cells and reduced immunosuppressive cells, thereby improving treatment efficacy. Additionally, induction of immunoactivation by an EGFR antibody enhances the antitumor effect of immune checkpoint inhibitors.
Inflammatory breast cancer (IBC), the most aggressive breast cancer subtype, is driven by an immunosuppressive tumor microenvironment (TME). Current treatments for IBC have limited efficacy. In a clinical trial (NCT01036087), an anti-EGFR antibody combined with neoadjuvant chemotherapy produced the highest pathological complete response rate ever reported in patients with IBC having triple-negative receptor status. We determined the molecular and immunological mechanisms behind this superior clinical outcome. Using novel humanized IBC mouse models, we discovered that EGFR-targeted therapy remodels the IBC TME by increasing cytotoxic T cells and reducing immunosuppressive regulatory T cells and M2 macrophages. These changes were due to diminishing immunosuppressive chemokine expression regulated by transcription factor EGR1. We also showed that induction of an immunoactive IBC TME by an anti-EGFR antibody improved the antitumor efficacy of an anti- PD-L1 antibody. Our findings lay the foundation for clinical trials evaluating EGFR-targeted therapy combined with immune checkpoint inhibitors in patients with cancer.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available