Oops, you're using an old version of your browser so some of the features on this page may not be displaying properly.

MINIMAL Requirements: Google Chrome 24+Mozilla Firefox 20+Internet Explorer 11Opera 15–18Apple Safari 7SeaMonkey 2.15-2.23

ePoster Display

1771P - Identification and validation of tissue or ctDNA PTPRD phosphatase domains deleterious mutations as prognostic and predictive biomarkers for ICIs in non-squamous NSCLC

Date

16 Sep 2021

Session

ePoster Display

Topics

Tumour Immunology;  Targeted Therapy;  Immunotherapy;  Translational Research

Tumour Site

Non-Small Cell Lung Cancer

Presenters

Yiting Sun

Citation

Annals of Oncology (2021) 32 (suppl_5): S1211-S1226. 10.1016/annonc/annonc716

Authors

Y. Sun1, J. Duan1, W. Fang2, Z. Wang1, L. Zhang2, H. Bai1, J. Wang1

Author affiliations

  • 1 Department Of Medical Oncology, Chinese Academy of Medical Sciences and Peking Union Medical College - National Cancer Center, Cancer Hospital, 100021 - Beijing/CN
  • 2 Medical Oncology, Sun Yat-sen University Cancer Center, 50017 - Guangzhou/CN

Resources

Login to get immediate access to this content.

If you do not have an ESMO account, please create one for free.

Abstract 1771P

Background

With the revolutionary progress of immune checkpoint inhibitors (ICIs) achieved in NSCLC, identifying patients benefiting from ICIs becomes critical and urgent. The associations of genomic alterations in protein tyrosine phosphatases receptor-type (PTPRs) and ICIs responses are unknown.

Methods

Whole-exome sequencing (WES) of 73 advanced NSCLC tumors sampled before anti-PD-(L)1therapy were carried out with corresponding clinical data collected as a discovery cohort. Three cohort of 1920 NSCLC patients with WES or target sequencing data of tumor tissue derived DNA or ctDNA and relevant clinical data were used to validate. The LUAD cohort in TCGA was used for analyzing the potential anti-tumor immunologic mechanisms.

Results

Among all PTPRs, PTRPD mutation in non-squamous NSCLC (ns-NSCLC) patients brought longer PFS (324 vs 63 days, HR=0.36, p= 0.015) and higher ORR (p=0.010) in discovery cohort. In validation cohort-1 (n=377), ns-NSCLC patients with tissue PTPRD mutations had favorable PFS (9.10 vs 4.33 months, HR=0.62, p=0.018) and ORR (p=0.013). In validation cohort-2 (n=406), ns-NSCLC patients with tissue PTPRD mutations had favorable OS (over 40 vs 11.94 months, HR=0.57, p=0.011). In validation cohort-3 (n=1137), ns-NSCLC patients with ctDNA PTPRD mutations had longer PFS (6.97 vs 2.73 months, HR=0.63, p=0.028) and higher ORR (p=0.047). Moreover, it was deleterious mutations in phosphatase domains (phosphatase-mut), rather than other mutations (other-mut), that were responsible of PTPRD’s prediction efficiency. In addition, in validation cohort-3, ctDNA phosphatase-mut was also a predictive biomarker identifying patients benefiting more from ICIs than chemotherapy (interaction P for PFS=0.0506, for OS=0.04). Univariate and multivariate regression analysis revealed phosphatase-mut was independent on PD-L1 expression and TMB to predict. In silico analysis based on TCGA LUAD cohort discovered enhanced anti-tumor immunity in phosphatase-mut patients.

Conclusions

Tissue or ctDNA PTPRD phosphatase domain deleterious mutations functioned as a both prognostic and predictive biomarker predicting clinical outcomes of ICIs in ns-NSCLC patients.

Clinical trial identification

Editorial acknowledgement

Legal entity responsible for the study

The authors.

Funding

This work was supported by National Key Research and Development Project 2019YFC1315700; NSFC Key Program [81630071]; NSFC General Program (81871889, 81972905); AiyouFoundation (KY201701).

Disclosure

All authors have declared no conflicts of interest.

This site uses cookies. Some of these cookies are essential, while others help us improve your experience by providing insights into how the site is being used.

For more detailed information on the cookies we use, please check our Privacy Policy.

Customise settings
  • Necessary cookies enable core functionality. The website cannot function properly without these cookies, and you can only disable them by changing your browser preferences.