4.7 Article

Recurrent Human Papillomavirus-Related Head and Neck Cancer Undergoes Metabolic Reprogramming and Is Driven by Oxidative Phosphorylation

Journal

CLINICAL CANCER RESEARCH
Volume 27, Issue 22, Pages 6250-6264

Publisher

AMER ASSOC CANCER RESEARCH

Keywords

-

Categories

Funding

  1. Department of Veterans Affairs [I01 BX-003456]
  2. NIH [RO1-DE028343, R00CA207871]
  3. Myers Family Foundation
  4. Mosites Personalized Medicine Fund

Ask authors/readers for more resources

HPV infection drives the development of some head and neck squamous cell carcinomas (HNSCC), with mechanisms underlying treatment failure still unclear. The OXPHOS pathway is enriched in recurrent HPV-associated HNSCC, potentially contributing to treatment failure. NRF2 plays a crucial role in HPV-related HNSCC and could be targeted for therapeutic intervention through metabolic reprogramming.
Purpose: Human papillomavirus (HPV) infection drives the development of some head and neck squamous cell carcinomas (HNSCC). This disease is rapidly increasing in incidence worldwide. Although these tumors are sensitive to treatment, approximately 10% of patients fail therapy. However, the mechanisms that underlie treatment failure remain unclear. Experimental Design: We performed RNA sequencing (RNA-seq) on tissues from matched primary-(pHNSCC) and metachronous-recurrent cancers (rHNSCC) to identify transcriptional differences to gain mechanistic insight into the evolutionary adaptations of metachronous-recurrent tumors. We used HPV-related HNSCC cells lines to investigate the effect of (i) NRF2 overexpression on growth in vitro and in vivo, (ii) oxidative phosphorylation (OXPHOS) inhibition using IACS-010759 on NRF2-dependent cells, and (iii) combination of cisplatin and OXPHOS inhibition. Results: The OXPHOS pathway is enriched in recurrent HPV-associated HNSCC and may contribute to treatment failure. NRF2-enriched HNSCC samples from The Cancer Genome Atlas (TCGA) with enrichment in OXPHOS, fatty-acid metabolism, Myc, Mtor, reactive oxygen species (ROS), and glycolytic signaling networks exhibited worse survival. HPV-positive HNSCC cells demonstrated sensitivity to the OXPHOS inhibitor, in a NRF2-dependent manner. Further, using murine xenograft models, we identified NRF2 as a driver of tumor growth. Mechanistically, NRF2 drives ROS and mitochondrial respiration, and NRF2 is a critical regulator of redox homeostasis that can be crippled by disruption of OXPHOS. NRF2 also mediated cisplatin sensitivity in endogenously overexpressing primary HPV-related HNSCC cells. Conclusions: These results unveil a paradigm-shifting translational target harnessing NRF2-mediated metabolic reprogramming in HPV-related HNSCC.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available