IL6 Blockade Reprograms the Lung Tumor Microenvironment to Limit the Development and Progression of K-ras–Mutant Lung Cancer

Autor: Amber M. Cumpian, Soudabeh Daliri, Seyed Javad Moghaddam, Mauricio S. Caetano, Stephanie S. Watowich, Cesar E. Ochoa, Nese Unver, Humam Kadara, Huiyuan Zhang, Seon Hee Chang, Lei Gong, Carmen Behrens, Samir M. Hanash, Carlos Gil Ferreira, Cinthya Sternberg, Edwin J. Ostrin, Ignacio I. Wistuba
Rok vydání: 2016
Předmět:
0301 basic medicine
Cancer Research
Lung Neoplasms
Carcinogenesis
Angiogenesis
Apoptosis
medicine.disease_cause
Immunoenzyme Techniques
Mice
Pulmonary Disease
Chronic Obstructive

0302 clinical medicine
Carcinoma
Non-Small-Cell Lung

Tumor Cells
Cultured

Tumor Microenvironment
Reverse Transcriptase Polymerase Chain Reaction
NF-kappa B
Antibodies
Monoclonal

Prognosis
Survival Rate
medicine.anatomical_structure
Oncology
030220 oncology & carcinogenesis
Disease Progression
Adenocarcinoma
Signal Transduction
STAT3 Transcription Factor
Blotting
Western

Biology
Real-Time Polymerase Chain Reaction
Article
Proto-Oncogene Proteins p21(ras)
03 medical and health sciences
Immune system
medicine
Animals
Humans
RNA
Messenger

Lung cancer
Cell Proliferation
Neoplasm Staging
Tumor microenvironment
Lung
Interleukin-6
Cancer
medicine.disease
Disease Models
Animal

030104 developmental biology
Mutation
Immunology
Zdroj: Cancer Research. 76:3189-3199
ISSN: 1538-7445
0008-5472
Popis: Activating mutations of K-ras are the most common oncogenic alterations found in lung cancer. Unfortunately, attempts to target K-ras–mutant lung tumors have thus far failed, clearly indicating the need for new approaches in patients with this molecular profile. We have previously shown NF-κB activation, release of IL6, and activation of its responsive transcription factor STAT3 in K-ras–mutant lung tumors, which was further amplified by the tumor-enhancing effect of chronic obstructive pulmonary disease (COPD)-type airway inflammation. These findings suggest an essential role for this inflammatory pathway in K-ras–mutant lung tumorigenesis and its enhancement by COPD. Therefore, here we blocked IL6 using a monoclonal anti-IL6 antibody in a K-ras–mutant mouse model of lung cancer in the absence or presence of COPD-type airway inflammation. IL6 blockade significantly inhibited lung cancer promotion, tumor cell–intrinsic STAT3 activation, tumor cell proliferation, and angiogenesis markers. Moreover, IL6 inhibition reduced expression of protumor type 2 molecules (arginase 1, Fizz 1, Mgl, and IDO), number of M2-type macrophages and granulocytic myeloid-derived suppressor cells, and protumor T-regulatory/Th17 cell responses. This was accompanied by increased expression of antitumor type 1 molecule (Nos2), and antitumor Th1/CD8 T-cell responses. Our study demonstrates that IL6 blockade not only has direct intrinsic inhibitory effect on tumor cells, but also reeducates the lung microenvironment toward an antitumor phenotype by altering the relative proportion between protumor and antitumor immune cells. This information introduces IL6 as a potential druggable target for prevention and treatment of K-ras–mutant lung tumors. Cancer Res; 76(11); 3189–99. ©2016 AACR.
Databáze: OpenAIRE