Radiomic signatures reveal multiscale intratumor heterogeneity associated with tissue tolerance and survival in re-irradiated nasopharyngeal carcinoma: a multicenter study

Post-radiation nasopharyngeal necrosis (PRNN) is a severe adverse event following re-radiotherapy for patients with locally recurrent nasopharyngeal carcinoma (LRNPC) and associated with decreased survival. Biological heterogeneity in recurrent tumors contributes to the different risks of PRNN. Radi...

Full description

Saved in:
Bibliographic Details
Published inBMC medicine Vol. 21; no. 1; p. 464
Main Authors Liu, Ting, Dong, Di, Zhao, Xun, Ou, Xiao-Min, Yi, Jun-Lin, Guan, Jian, Zhang, Ye, Xiao-Fei, Lv, Xie, Chuan-Miao, Luo, Dong-Hua, Sun, Rui, Chen, Qiu-Yan, Xing, Lv, Guo, Shan-Shan, Liu, Li-Ting, Lin, Da-Feng, Chen, Yan-Zhou, Lin, Jie-Yi, Luo, Mei-Juan, Yan, Wen-Bin, He, Mei-Lin, Mao, Meng-Yuan, Zhu, Man-Yi, Chen, Wen-Hui, Shen, Bo-Wen, Wang, Shi-Qian, Li, Hai-Lin, Zhong, Lian-Zhen, Hu, Chao-Su, Wu, De-Hua, Mai, Hai-Qiang, Tian, Jie, Tang, Lin-Quan
Format Journal Article
LanguageEnglish
Published England BioMed Central Ltd 27.11.2023
BioMed Central
BMC
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Post-radiation nasopharyngeal necrosis (PRNN) is a severe adverse event following re-radiotherapy for patients with locally recurrent nasopharyngeal carcinoma (LRNPC) and associated with decreased survival. Biological heterogeneity in recurrent tumors contributes to the different risks of PRNN. Radiomics can be used to mine high-throughput non-invasive image features to predict clinical outcomes and capture underlying biological functions. We aimed to develop a radiogenomic signature for the pre-treatment prediction of PRNN to guide re-radiotherapy in patients with LRNPC. This multicenter study included 761 re-irradiated patients with LRNPC at four centers in NPC endemic area and divided them into training, internal validation, and external validation cohorts. We built a machine learning (random forest) radiomic signature based on the pre-treatment multiparametric magnetic resonance images for predicting PRNN following re-radiotherapy. We comprehensively assessed the performance of the radiomic signature. Transcriptomic sequencing and gene set enrichment analyses were conducted to identify the associated biological processes. The radiomic signature showed discrimination of 1-year PRNN in the training, internal validation, and external validation cohorts (area under the curve (AUC) 0.713-0.756). Stratified by a cutoff score of 0.735, patients with high-risk signature had higher incidences of PRNN than patients with low-risk signature (1-year PRNN rates 42.2-62.5% vs. 16.3-18.8%, P < 0.001). The signature significantly outperformed the clinical model (P < 0.05) and was generalizable across different centers, imaging parameters, and patient subgroups. The radiomic signature had prognostic value concerning its correlation with PRNN-related deaths (hazard ratio (HR) 3.07-6.75, P < 0.001) and all causes of deaths (HR 1.53-2.30, P < 0.01). Radiogenomics analyses revealed associations between the radiomic signature and signaling pathways involved in tissue fibrosis and vascularity. We present a radiomic signature for the individualized risk assessment of PRNN following re-radiotherapy, which may serve as a noninvasive radio-biomarker of radiation injury-associated processes and a useful clinical tool to personalize treatment recommendations for patients with LANPC.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:1741-7015
1741-7015
DOI:10.1186/s12916-023-03164-3