Abstract

Simple SummaryImmune checkpoint blockade with anti-PD-1 antibodies blocks the development of oral squamous cell carcinomas (OSCCs) in preclinical models. Understanding whether the genetic alterations that accumulate during oral cancer development affect the response to PD-1 inhibitors is critical to identify patients who may benefit from immunoprevention interventions. Using genetically engineered mouse models that develop carcinogen-induced oral tumors that differ on the mutational status of the p53 gene, we demonstrated that expression of gain-of-function mutant p53 in the epithelial cells of the oral lesions promotes resistance to the immunopreventive effects of anti-PD-1. These novel findings could guide patient-specific strategies for oral cancer immunoprevention based on p53 profiling.Oral squamous cell carcinoma (OSCC) develops through the multistep malignant progression of squamous epithelium. This process can be prevented by PD-1 blockade in a mouse model for oral carcinogenesis. OSCCs exhibit a high incidence of p53 mutations that confer oncogenic gain-of-function (GOF) activities that promote resistance to standard therapies and poor clinical outcomes. To determine whether epithelial p53 mutations modulate anti-PD-1-mediated oral cancer immunoprevention, we generated mouse models for oral carcinogenesis by exposing mice carrying epithelial-specific p53 mutations to the carcinogen 4NQO. Consistent with the oncogenic functions of mutant p53, mice with OSCCs expressing the p53R172H GOF mutation developed higher metastasis rates than mice with loss-of-function (LOF) p53 deletion or with wild-type p53. Throughout oral cancer progression, pre-invasive and invasive lesions showed a gradual increase in T-cell infiltration, recruitment of immunosuppressive regulatory T-cells (Tregs), and induction of PD-1/PD-L1 immune checkpoint proteins. Notably, while PD-1 blockade prevented the development of OSCCs in mice with wild-type p53 or p53 deletion, GOF p53R172H abrogated the immunopreventive effects of anti-PD-1, associated with upregulation of IL17 signaling and depletion of exhausted CD8 cells in the microenvironment of the p53R172H tumors. These findings sustain a potential role for p53 profiling in personalized oral cancer immunoprevention.

Highlights

  • Oral cancer is the most frequent malignancy of the head and neck and the sixth most common human cancer worldwide, with five-year survival rates that remain at 50% [1,2].Over 90% of the oral cancers are squamous cell carcinomas (OSCCs) that develop from the oral squamous epithelium following a multistep process of malignant progression that involves the acquisition of genetic and phenotypic alterations promoted by tobacco and/or alcohol as the main etiological factors [3,4]

  • To determine the contribution of p53 mutations to oral cancer metastasis, mice were monitored until development of morbidity endpoints according to Institutional Animal Care and Use Committee (IACUC) regulations

  • The findings demonstrate a gradual recruitment of immune infiltrates and PD-1/PD-L1 immune checkpoint activation during oral carcinogenesis and suggests that p53 mutations may alter this dynamic process

Read more

Summary

Introduction

Oral cancer is the most frequent malignancy of the head and neck and the sixth most common human cancer worldwide, with five-year survival rates that remain at 50% [1,2]. Over 90% of the oral cancers are squamous cell carcinomas (OSCCs) that develop from the oral squamous epithelium following a multistep process of malignant progression that involves the acquisition of genetic and phenotypic alterations promoted by tobacco and/or alcohol as the main etiological factors [3,4]. Recent clinical trials with blocking antibodies for the immune checkpoint molecule programmed cell death protein 1 (PD-1) have shown to improve survival in patients with advanced head and neck squamous cell carcinoma [11,12,13]. Our previous studies showed that PD-1 blockade can prevent the malignant progression of OPLs, in a mouse model for oral carcinogenesis, supporting the preventive potential of immune checkpoint inhibition to contain oral cancer development [14]

Objectives
Methods
Results
Conclusion
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