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

691P - Effect of NANOGNB mutations on DNA damage response and efficacy of immune checkpoint inhibitors in advanced renal clear cell carcinoma

Date

16 Sep 2021

Session

ePoster Display

Topics

Tumour Site

Renal Cell Cancer

Presenters

Zhiwen Luo

Citation

Annals of Oncology (2021) 32 (suppl_5): S678-S724. 10.1016/annonc/annonc675

Authors

Z. Luo1, X. Bi1, X. Bi2

Author affiliations

  • 1 Department Of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021 - Beijing/CN
  • 2 Department Of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021 - Beijing/CN

Resources

Login to get immediate access to this content.

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

Abstract 691P

Background

Immune checkpoint inhibitors (ICIs) have achieved impressive success in advanced renal clear cell carcinoma (ccRCC), yet responses vary and predictive biomarkers are urgently needed. Growing evidence has suggested that specific genetic mutations may serve as immune biomarkers for immune checkpoint blockade therapy of many solid cancer.

Methods

Clinical ICIs-treated cohorts of advanced ccRCC with annotated response and survival data and matched mutational data were collected and consolidated. The predictive function of all mutated genes was first tested in the discovery cohort and later validated in the validation cohort. The association between mutated genes and tumor immunogenicity and anti-tumor immunity was further investigated.

Results

In the discovery cohort, among 12379 genes in 245 advanced ccRCC treated with ICIs, NANOGNB-mutant (NANOGNB-MUT) was enriched in patients responding to ICI treatment (P<0.001). Compared with other responsive related gene mutations, NANOGNB was mutated in largest proportion (5% advanced ccRCC). Significant differences were observed between NANOGNB-MUT and NANOGNB-wildtype (NANOGNB-WT) regarding objective response rate (ORR, 54.55% versus 21.37%, P<0.001), durable clinical benefit (DCB, 63.64% versus 30.34%, P<0.001), progression-free survival (PFS, hazard ratio=0.45 [95% confidence interval, 0.22 to 0.92], P=0.022), which was, importantly, independent of tumor mutational burden and high microsatellite instability; as well as not attributed to the prognostic impact of NANOGNB-MUT (P>0.05 in non-ICI-treated cohorts, 280 advanced ccRCCs). NANOGNB-MUT was associated with known predictors of ICB therapy efficacy, such as higher tumor mutational burden (TMB), neoantigen load and the number of mutations in the DNA damage-repair pathway. In addition, NANOGNB-MUT tumors had damaged DNA damage response, highly infiltrating TILs, and relatively activated antitumor immunity, suggesting that NANOGNB mutations may serve as a potential biomarker to guide ICB therapy in advanced ccRCCs.

Conclusions

NANOGNB-MUT was strongly associated with higher ORR, better DCB, longer PFS in advanced ccRCCs in ICIs, suggesting that NANOGNB-MUT is a novel predictive biomarker for ICIs for ccRCCs.

Clinical trial identification

Editorial acknowledgement

Legal entity responsible for the study

Zhiwen Luo, Xinyu Bi and Xingang Bi.

Funding

Has not received any funding.

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.