Lipopolysaccharide-Mediated Chronic Inflammation Promotes Tobacco Carcinogen-Induced Lung Cancer and Determines the Efficacy of Immunotherapy

Cancer Res. 2021 Jan 1;81(1):144-157. doi: 10.1158/0008-5472.CAN-20-1994. Epub 2020 Oct 29.

Abstract

Chronic obstructive pulmonary disease (COPD) is an inflammatory disease that is associated with increased risk of lung cancer. Pseudomonas aeruginosa (PA) infections are frequent in patients with COPD, which increase lung inflammation and acute exacerbations. However, the influences of PA-induced inflammation on lung tumorigenesis and the efficacy of immune checkpoint blockade remain unknown. In this study, we initiated a murine model of lung cancer by treating FVB/NJ female mice with tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) alone or in combination with PA-lipopolysaccharide (LPS). LPS-mediated chronic inflammation induced T-cell exhaustion, increased the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis, and enhanced NNK-induced lung tumorigenesis through an immunosuppressive microenvironment characterized by accumulation of myeloid-derived suppressive cells (MDSC) and regulatory T cells. Anti-PD-1 antibody treatment reduced tumors in NNK/LPS-treated mice with a 10-week LPS treatment but failed to inhibit tumor growth when LPS exposure was prolonged to 16 weeks. Anti-Ly6G antibody treatment coupled with depletion of MDSC alone reduced tumor growth; when combined with anti-PD-1 antibody, this treatment further enhanced antitumor activity in 16-week NNK/LPS-treated mice. Immune gene signatures from a human lung cancer dataset of PD-1 blockade were identified, which predicted treatment responses and survival outcome and overlapped with those from the mouse model. This study demonstrated that LPS-mediated chronic inflammation creates a favorable immunosuppressive microenvironment for tumor progression and correlates with the efficacy of anti-PD-1 treatment in mice. Immune gene signatures overlap with human and mouse lung tumors, providing potentially predictive markers for patients undergoing immunotherapy. SIGNIFICANCE: This study identifies an immune gene signature that predicts treatment responses and survival in patients with tobacco carcinogen-induced lung cancer receiving immune checkpoint blockade therapy.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, N.I.H., Intramural

MeSH terms

  • Animals
  • Biomarkers, Tumor / genetics
  • Biomarkers, Tumor / metabolism
  • Butanones / toxicity*
  • Carcinogens / toxicity*
  • Female
  • Gene Expression Profiling
  • Gene Expression Regulation, Neoplastic
  • Humans
  • Immune Checkpoint Inhibitors / pharmacology*
  • Inflammation / chemically induced
  • Inflammation / complications*
  • Lipopolysaccharides / toxicity*
  • Lung Neoplasms / drug therapy
  • Lung Neoplasms / etiology
  • Lung Neoplasms / metabolism
  • Lung Neoplasms / pathology*
  • Mice
  • Nicotiana / toxicity*
  • Nitrosamines / toxicity*
  • Prognosis
  • Programmed Cell Death 1 Receptor / antagonists & inhibitors
  • Survival Rate
  • Tumor Microenvironment / drug effects
  • Tumor Microenvironment / immunology

Substances

  • Biomarkers, Tumor
  • Butanones
  • Carcinogens
  • Immune Checkpoint Inhibitors
  • Lipopolysaccharides
  • Nitrosamines
  • Pdcd1 protein, mouse
  • Programmed Cell Death 1 Receptor
  • nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone