1. Academic Validation
  2. Interplay between estrogen and Stat3/NF-κB-driven immunomodulation in lung cancer

Interplay between estrogen and Stat3/NF-κB-driven immunomodulation in lung cancer

  • Carcinogenesis. 2020 Nov 13;41(11):1529-1542. doi: 10.1093/carcin/bgaa064.
Shanshan Deng 1 2 Marco Ramos-Castaneda 2 Walter V Velasco 2 Michael J Clowers 2 3 Berenice A Gutierrez 2 Oscar Noble 2 Yiping Dong 4 Melody Zarghooni 2 Lucero Alvarado 5 Mauricio S Caetano 2 Shuanying Yang 1 Edwin J Ostrin 5 6 Carmen Behrens 7 Ignacio I Wistuba 8 Laura P Stabile 9 Humam Kadara 3 8 Stephanie S Watowich 3 10 Seyed Javad Moghaddam 2 3
Affiliations

Affiliations

  • 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
  • 2 Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
  • 3 The University of Texas M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
  • 4 Department of Oncology Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
  • 5 Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
  • 6 Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
  • 7 Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
  • 8 Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
  • 9 Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
  • 10 Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Abstract

K-Ras mutant lung adenocarcinoma (LUAD) is the most common type of lung Cancer, displays abysmal prognosis and is tightly linked to tumor-promoting inflammation, which is increasingly recognized as a target for therapeutic intervention. We have recently shown a gender-specific role for epithelial STAT3 signaling in the pathogenesis of K-Ras mutant LUAD. The absence of epithelial STAT3 in male K-Ras mutant mice (LR/Stat3Δ/Δ mice) promoted tumorigenesis and induced a nuclear factor-kappaB (NF-κB)-driven pro-tumor immune response while reducing tumorigenesis and enhancing anti-tumor immunity in female counterparts. In the present study, we manipulated estrogen and NF-κB signaling to study the mechanisms underlying this intriguing gender-disparity. In LR/Stat3Δ/Δ females, estrogen deprivation by bilateral oophorectomy resulted in higher tumor burden, an induction of NF-κB-driven immunosuppressive response, and reduced anti-tumor cytotoxicity, whereas estrogen replacement reversed these changes. On the other hand, exogenous estrogen in males successfully inhibited tumorigenesis, attenuated NF-κB-driven immunosuppression and boosted anti-tumor immunity. Mechanistically, genetic targeting of epithelial NF-κB activity resulted in reduced tumorigenesis and enhanced the anti-tumor immune response in LR/Stat3Δ/Δ males, but not females. Our data suggest that estrogen exerts a context-specific anti-tumor effect through inhibiting NF-κB-driven tumor-promoting inflammation and provide insights into developing novel personalized therapeutic strategies for K-Ras mutant LUAD.

Figures
Products
  • Cat. No.
    Product Name
    Description
    Target
    Research Area
  • HY-10172
    99.51%, IKKβ Inhibitor
    IKK