Poster Presentation 31st Lorne Cancer Conference 2019

Distinct cancer-promoting stromal gene expression programs depending on lung function (#318)

Brian J Sandri 1 , Laia Masvidal 2 , Carl Murie 2 , Margarita Bartish 2 , Svetlana Avdulov 1 , LeeAnn Higgins 3 , Todd Markowski 3 , Mark Peterson 1 , Jonas Bergh 2 , Ping Yang 4 , Charlotte Rolny 2 , Andrew H Limper 5 , Timothy J Griffin 3 , Peter B Bitterman 1 , Chris H Wendt 1 , Ola Larsson 2
  1. Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
  2. Karolinska Institute, Solna, STOCKHOLM, Sweden
  3. Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
  4. Department of Epidemiology, Mayo Clinic, Rochester, MN, USA
  5. Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA

Rationale: Chronic obstructive pulmonary disease (COPD) is an independent risk factor for lung cancer, but the underlying molecular mechanisms are unknown.  We hypothesized that lung stromal cells activate pathological gene expression programs supporting oncogenesis.

Objective: To identify molecular mechanisms operating in the lung stroma that support development of lung cancer.

Methods: Study subjects included patients with- or without- lung cancer across a spectrum of lung function. We conducted multi-omics analysis of non-malignant lung tissue to quantify the transcriptome, translatome and proteome.

Measurements and Main Results: Cancer-associated gene expression changes predominantly manifested as alterations in the efficiency of mRNA translation modulating protein levels in the absence of corresponding changes in mRNA levels. The molecular mechanisms driving these cancer-associated translation programs differed based on lung function. In subjects with normal to mildly impaired lung-function, the mammalian target of rapamycin (mTOR) pathway served as an upstream driver; whereas in severe airflow obstruction, pathways downstream of pathological extracellular matrix (ECM) emerged. Consistent with a role during cancer initiation, both the mTOR and ECM gene expression programs paralleled activation of previously identified pro-cancer secretomes. Furthermore, in situ examination of lung tissue documented that stromal fibroblasts express cancer-associated proteins from the two pro-cancer secretomes including IL6 in mild airflow obstruction and BMP1 in severe airflow obstruction.

Conclusion: Two distinct stromal gene expression programs promoting cancer initiation are activated in lung cancer patients depending on lung function. Our work has implications both for screening strategies and personalized approaches to cancer treatment.