STING Pathway Expression Identifies NSCLC With an Immune-Responsive Phenotype

Carminia M. Della Corte, Triparna Sen, Carl M. Gay, Kavya Ramkumar, Lixia Diao, Robert J. Cardnell, Bertha Leticia Rodriguez, C. Allison Stewart, Vassiliki A. Papadimitrakopoulou, Laura Gibson, Jared J. Fradette, Qi Wang, Youhong Fan, David H. Peng, Marcelo V. Negrao, Ignacio I. Wistuba, Junya Fujimoto, Luisa M. Solis Soto, Carmen Behrens, Ferdinandos SkoulidisJohn V. Heymach, Jing Wang, Don L. Gibbons, Lauren A. Byers

Research output: Contribution to journalArticlepeer-review

82 Scopus citations

Abstract

Introduction: Although the combination of anti–programmed cell death-1 or anti–programmed cell death ligand-1 (PD-L1) with platinum chemotherapy is a standard of care for NSCLC, clinical responses vary. Even though predictive biomarkers (which include PD-L1 expression, tumor mutational burden, and inflamed immune microenvironment) are validated for immunotherapy, their relevance to chemoimmunotherapy combinations is less clear. We have recently reported that activation of the stimulator of interferon genes (STING) innate immune pathway enhances immunotherapy response in SCLC. Here, we hypothesize that STING pathway activation may predict and underlie predictive correlates of antitumor immunity in NSCLC. Methods: We analyzed transcriptomic and proteomic profiles in two NSCLC cohorts from our institution (treatment-naive patients in the Profiling of Resistance Patterns and Oncogenic Signaling Pathways in Evaluation of Cancers of the Thorax study and relapsed patients in the Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination study) and The Cancer Genome Atlas (N = 1320). Tumors were stratified by STING activation on the basis of protein or mRNA expression of cyclic GMP-AMP synthase, phospho-STING, and STING-mediated chemokines (chemokine ligand 5 [CCL5] and C-X-C motif chemokine 10 [CXCL10]). STING activation in patient tumors and in platinum-treated preclinical NSCLC models was correlated with biomarkers of immunotherapy response. Results: STING activation is associated with higher levels of intrinsic DNA damage, targetable immune checkpoints, and chemokines in treatment-naive and relapsed lung adenocarcinoma. We observed that tumors with lower STING and immune gene expression show higher frequency of serine-threonine kinase 11 (STK11) mutations; however, we identified a subset of these tumors that are TP53 comutated and display high immune- and STING-related gene expression. Treatment with cisplatin increases STING pathway activation and PD-L1 expression in multiple NSCLC preclinical models, including adeno- and squamous cell carcinoma. Conclusions: STING pathway activation in NSCLC predicts features of immunotherapy response and is enhanced by cisplatin treatment. This suggests a possible predictive biomarker and mechanism for improved response to chemoimmunotherapy combinations.

Original languageEnglish (US)
Pages (from-to)777-791
Number of pages15
JournalJournal of Thoracic Oncology
Volume15
Issue number5
DOIs
StatePublished - May 2020

Keywords

  • Immune checkpoints
  • Immunotherapy
  • Innate immunity
  • Lung cancer
  • STING

ASJC Scopus subject areas

  • Oncology
  • Pulmonary and Respiratory Medicine

MD Anderson CCSG core facilities

  • Bioinformatics Shared Resource
  • Functional Proteomics Reverse Phase Protein Array Core
  • Tissue Biospecimen and Pathology Resource
  • Advanced Technology Genomics Core
  • Cytogenetics and Cell Authentication Core
  • Research Animal Support Facility

Fingerprint

Dive into the research topics of 'STING Pathway Expression Identifies NSCLC With an Immune-Responsive Phenotype'. Together they form a unique fingerprint.

Cite this