NOTCH4 mutation as a potential predictive biomarker for immunotherapy in NRAS wildtype melanoma.

Authors

null

Hongxia Li

Shanxi Provincial People's Hospital, Taiyuan City, China

Hongxia Li , Qin Zhang , Qianqian Duan , Yuan Tan , Tingting Sun , Chuang Qi , Yong Ren

Organizations

Shanxi Provincial People's Hospital, Taiyuan City, China, The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd; Nanjing Simcere Medical Laboratory Science Co., Ltd; The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China, The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China, The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China

Research Funding

No funding received

Background: NRAS wildtype melanoma account for 80-85% melanoma patients and have been reported had a better response for immunotherapy. Although PD-L1 expression and tumor mutation burden (TMB) have been considered as important biomarkers for immunotherapy in many types of tumors, the predictive efficacy of them in melanoma is controversial. Since evidences showed NOTCH pathway mutation was a potential predictive biomarker for immune checkpoint inhibitors in some types of tumors. Here we explored the relationship between NOTCH family genes mutations and efficacy of NRAS wildtype melanoma immunotherapy. Methods: 265 NRAS wildtype ICI-pretreatment melanomas from cBioPortal (https://www.cbioportal.org), which is divided to discovery (N = 76) and validation (N = 189) cohort to analyze the association between NOTCH family genes (NOTCH1-4) mutation and efficacy of ICIs therapy. All nonsynonymous somatic mutations, including nonsense, missense, nonstop, frameshift deletion and insertion, in-frame deletion and insertion, and splice site mutations were considered for inclusion in our study. The potential mechanism was subsequently explored through RNA expression in immunotherapy treated population. Results: Patients with NOTCH4-Mut were identified to be associated with prolonged overall survival (OS) in discovery (HR: 0.30, 95%CI: 0.11-0.83, P = 0.01) and validation cohort (HR: 0.21, 95%CI: 0.07-0.68, P = 0.003). Further exploration found that NOTCH4-Mut tumors had higher tumor mutation burden (TMB) and tumor neoantigen burden (TNB) (P < 0.05). RNA data revealed that enhanced anti-tumor immunity was observed in NOTCH4-Mut tumors. Activated CD4 memory T cell, CD8 T cell, follicular helper T cell (Tfh) and Neutrophils were more abundant in NOTCH4-Mut tumors. In addition, the results of enrichment analysis showed that several immune-related pathways varied significantly between NOTCH4-Mut and NOTCH4-Wt tumors, including Interferon γ response, DNA repair, epithelial mesenchymal transition (EMT), angiogenesis. Conclusions: NOTCH4 mutation may promote tumor immunity and serve as a biomarker to predict good immune response in NRAS wildtype melanoma and guiding immunotherapeutic responsiveness.

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2022 ASCO Annual Meeting

Session Type

Publication Only

Session Title

Melanoma/Skin Cancers

Track

Melanoma/Skin Cancers

Sub Track

Other Melanoma/Skin Cancers

Citation

J Clin Oncol 40, 2022 (suppl 16; abstr e21594)

DOI

10.1200/JCO.2022.40.16_suppl.e21594

Abstract #

e21594

Abstract Disclosures

Similar Abstracts

Abstract

2023 ASCO Gastrointestinal Cancers Symposium

Tumor mutational burden as a potential predictive marker for the efficacy of immunotherapy in advanced gastric cancer.

First Author: Jae Yeon Jang

Abstract

2023 ASCO Annual Meeting

A cilia-related marker predicting suitability for immunotherapy in melanoma.

First Author: Changbin Zhu

Abstract

2024 ASCO Gastrointestinal Cancers Symposium

The combination of CD8 and TIM3 expression to predict survival outcomes in hepatocellular carcinoma.

First Author: Mitsuo Shimada