Abstract

Abstract The NRF2/KEAP1 pathway regulates cytoprotective mechanisms that protect healthy cells from malignant transformation and maintain cellular homeostasis. Up to 30% of human lung tumors contain a mutation that results in constitutive NRF2 pathway activation, which contributes to cancer cell survival, disease progression, and resistance to chemotherapies. However, the effects of NRF2 activation in immune cells within the tumor microenvironment are underexplored. Macrophages in lung cancers contribute to cancer progression or regression depending on the context, and NRF2 activity affects macrophage activation and trafficking. To evaluate the role of NRF2 activity in macrophages, bone marrow-derived macrophages (BMDMs) from NRF2 wild-type (WT) and knockout (KO) mice were treated to induce anti-tumor (M1), tumor-promoting (M2), and tumor-educated macrophage phenotypes and then treated with the potent NRF2 activator CDDO-Methyl ester (CDDO-Me), also known as bardoxolone methyl. In tumor-educated BMDMs isolated from NRF2 WT mice and cultured in conditioned media from lung cancer cells, CDDO-Me (100 nM) induced a 3-fold increase in mRNA expression of the M1 markers TNF-α and IL-6. The expression of these cytokines was not increased by CDDO-Me in tumor-educated BMDMs isolated from NRF2 KO mice. CDDO-Me also decreased mRNA expression of the angiogenic factor VEGF and the chemokine CCL2 in tumor-educated BMDMs isolated from WT but not NRF2 KO mice. VEGF is expressed by tumor-promoting macrophages and CCL2, a chemotactic factor that recruits M2 macrophages into the lung, is a biomarker of poor prognosis in lung cancer patients. CDDO-Me has potent anti-tumor effects in the A/J mouse model of non-small cell lung cancer (NSCLC), but the mechanism of these effects was unknown. To evaluate, A/J WT and NRF2 KO A/J mice were injected with vinyl carbamate to initiate lung tumors. Two weeks later, mice were fed either a control diet or a diet containing CDDO-Me (50 mg/kg diet: ~12.5 mg/kg body weight) for 16 weeks. Ultrasound confirmed the development of tumors in the lungs of mice throughout the study, and differences in tumor number and lung parenchyma structure were detected at 15 weeks of treatment. CDDO-Me significantly (p<0.05) decreased tumor burden in WT A/J mice. Tumor burden in the NRF2 KO mice was significantly (p<0.05) higher than in WT mice, irrespective of treatment. Tumor count in NRF2 KO A/J mice treated with either vehicle or CDDO-Me was increased 2-fold compared to WT A/J mice on the vehicle control diet. These data show that CDDO-Me promotes an anti-cancer macrophage phenotype in vitro that is dependent on NRF2 activation. Additionally, NRF2 KO increases lung tumor burden in the A/J model of NSCLC and CDDO-Me significantly decreases tumor burden in A/J mice in a NRF2-dependent manner. Future studies will test the effect of CDDO-Me and NRF2 activity on the lung tumor immune microenvironment in vivo. Citation Format: Jess Ann Moerland, Karen T. Liby. The triterpenoid CDDO-Me redirects macrophage polarization and decreases tumor burden in a preclinical model of NSCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3534.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call