Glutathione peroxidase 2 is a metabolic driver of the tumor immune microenvironment and immune checkpoint inhibitor response.

TitleGlutathione peroxidase 2 is a metabolic driver of the tumor immune microenvironment and immune checkpoint inhibitor response.
Publication TypeJournal Article
Year of Publication2022
AuthorsAhmed, KMokim, Veeramachaneni, R, Deng, D, Putluri, N, Putluri, V, Cardenas, MF, Wheeler, DA, Decker, WK, Frederick, AI, Kazi, S, Sikora, AG, Sandulache, VC, Frederick, MJ
JournalJ Immunother Cancer
Volume10
Issue8
Date Published2022 Aug
ISSN2051-1426
KeywordsAnimals, Dinoprostone, Glutathione Peroxidase, Immune Checkpoint Inhibitors, Mice, NF-E2-Related Factor 2, Tumor Microenvironment
Abstract

BACKGROUND: The existence of immunologically 'cold tumors' frequently found across a wide spectrum of tumor types represents a significant challenge for cancer immunotherapy. Cold tumors have poor baseline pan-leukocyte infiltration, including a low prevalence of cytotoxic lymphocytes, and not surprisingly respond unfavorably to immune checkpoint (IC) inhibitors. We hypothesized that cold tumors harbor a mechanism of immune escape upstream and independent of ICs that may be driven by tumor biology rather than differences in mutational neoantigen burden.

METHODS: Using a bioinformatic approach to analyze TCGA (The Cancer Genome Atlas) RNA sequencing data we identified genes upregulated in cold versus hot tumors across four different smoking-related cancers, including squamous carcinomas from the oral cavity (OCSCC) and lung (LUSC), and adenocarcinomas of the bladder (BLCA) and lung (LUAD). Biological significance of the gene most robustly associated with a cold tumor phenotype across all four tumor types, glutathione peroxidase 2 (GPX2), was further evaluated using a combination of in silico analyses and functional genomic experiments performed both in vitro and in in vivo with preclinical models of oral cancer.

RESULTS: Elevated RNA expression of five metabolic enzymes including GPX2, aldo-keto reductase family 1 members AKR1C1, AKR1C3, and cytochrome monoxygenases (CP4F11 and CYP4F3) co-occurred in cold tumors across all four smoking-related cancers. These genes have all been linked to negative regulation of arachidonic acid metabolism-a well-established inflammatory pathway-and are also known downstream targets of the redox sensitive Nrf2 transcription factor pathway. In OCSCC, LUSC, and LUAD, GPX2 expression was highly correlated with Nrf2 activation signatures, also elevated in cold tumors. In BLCA, however, GPX2 correlated more strongly than Nrf2 signatures with decreased infiltration of multiple leukocyte subtypes. GPX2 inversely correlated with expression of multiple pro- inflammatory cytokines/chemokines and NF-kB activation in cell lines and knockdown of GPX2 led to increased secretion of prostaglandin E2 (PGE2) and interleukin-6. Conversely, GPX2 overexpression led to reduced PGE2 production in a murine OCSCC model (MOC1). GPX2 overexpressing MOC1 tumors had a more suppressive tumor immune microenvironment and responded less favorably to anti-cytotoxic T-lymphocytes-associated protein 4 IC therapy in mice.

CONCLUSION: GPX2 overexpression represents a novel potentially targetable effector of immune escape in cold tumors.

DOI10.1136/jitc-2022-004752
Alternate JournalJ Immunother Cancer
PubMed ID36002187
PubMed Central IDPMC9413193
Grant ListR01 AI127387 / AI / NIAID NIH HHS / United States
P30 CA125123 / CA / NCI NIH HHS / United States
R03 DE028858 / DE / NIDCR NIH HHS / United States
S10 RR024574 / RR / NCRR NIH HHS / United States
IK2 CX001953 / CX / CSRD VA / United States

Similar Publications