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

E-Poster Display

1128P - Germline variation in PDCD1 is associated with survival in metastatic melanoma after anti-PD-1 monotherapy

Date

17 Sep 2020

Session

E-Poster Display

Topics

Immunotherapy

Tumour Site

Melanoma

Presenters

Mirjam De With

Citation

Annals of Oncology (2020) 31 (suppl_4): S672-S710. 10.1016/annonc/annonc280

Authors

M. De With1, D.P. Hurkmans2, E. Oomen-de Hoop3, A. Lalouti3, S. Bins3, S. El Bouazzaoui4, R. Debets3, J.G. Aerts5, R.H.N. Van Schaik4, R.H. Mathijssen3, A.A.M. Van der Veldt6

Author affiliations

  • 1 Medical Oncology & Clinical Chemistry, Erasmus MC Cancer Institute, 3015 GD - Rotterdam/NL
  • 2 Medical Oncology & Pulmonology, Erasmus MC Cancer Institute, 3015 GD - Rotterdam/NL
  • 3 Medical Oncology, Erasmus MC Cancer Institute, 3015 GD - Rotterdam/NL
  • 4 Clinical Chemistry, Erasmus University Medical Center, 3015 GD - Rotterdam/NL
  • 5 Pulmonology, Erasmus University Medical Center, 3015 GD - Rotterdam/NL
  • 6 Medical Oncology & Radiology And Nuclear Medicine, Erasmus University Medical Center, 3015 GD - Rotterdam/NL

Resources

Login to get immediate access to this content.

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

Abstract 1128P

Background

Interaction of the programmed cell death-1 (PD-1) receptor with its ligands leads to downregulation of the T cell effector function and impairs antitumor immunity. Immune checkpoint inhibitors (ICIs) targeting PD-1 have proven efficacy in patients with several malignancies. Single nucleotide polymorphisms (SNPs) in genes related to the PD-1 axis likely affect T cell response upon anti-PD treatment. In fact, specific SNPs in the PDCD1 gene downregulate expression of PD-1 by T cells and are associated with decreased susceptibility to cancer. In this study, we investigated whether SNPs in genes related to the PD-1 axis are predictive of survival in metastatic melanoma patients treated with anti-PD-1 monotherapy.

Methods

Consecutive patients with metastatic melanoma who were treated either with nivolumab or pembrolizumab were included. Prospectively collected samples were genotyped for SNPs in HLAA (rs60131261), GZMB (rs8192917), IL-10 (rs3024493), IL2RA (rs2104286), IL-2RB (rs3218253), IFNG (rs2430561; rs2069705; rs2069718), PDCD1 (rs2227981), PTPN11 (rs2301756), and ZAP70 (rs13420683). Associations between SNPs and overall survival (OS) or progression free survival (PFS) were tested using Cox regression analysis. Associations with p ≤ 0.10 were tested multivariably and internally validated by bootstrapping.

Results

In total, 119 patients were included. The median follow-up was 2.6 years. Variation of PDCD1 (rs2227981) and GZMB (rs8192917) showed a trend towards OS (HR 2.03; 95%CI: 0.10-4.12; p 0.051 and HR 1.81; 95%CI: 0.95-3.36; p 0.060, respectively) and PFS (GZMB: HR 1.63; 95%CI: 1.00-2.67; p 0.051). After correction for prognostic factors (i.e. age, ECOG performance status and LDH), only variation of PDCD1 remained significantly associated with poorer OS (HR 2.37; 95%CI: 1.11-5.04; p 0.026), which was internally validated (Bias-corrected 95%CI: 1.04-6.25).

Conclusions

Patients with metastatic melanoma who have germline variation of PDCD1 have a significant poorer overall survival after anti-PD-1 monotherapy. These findings suggest that germline variations in immune-related genes are relevant for response and resistance to ICIs, which could be further explored in genome wide association studies.

Clinical trial identification

Editorial acknowledgement

Legal entity responsible for the study

Erasmus University Medical Center.

Funding

Has not received any funding.

Disclosure

J.G. Aerts: Honoraria (self): MSD; Honoraria (self): BMS; Honoraria (self): Boehringer Ingelheim; Honoraria (self), Licensing/Royalties: Amphera; Honoraria (self): Eli-Lilly; Honoraria (self): Takeda; Honoraria (self): Bayer; Honoraria (self): Roche; Honoraria (self): AstraZeneca. R.H. Mathijssen: Research grant/Funding (self): Astellas; Research grant/Funding (self): Bayer; Research grant/Funding (self): Boehringer-Ingelheim; Research grant/Funding (self): Cristal Therapeutics; Research grant/Funding (self), Licensing/Royalties: Pamgene; Research grant/Funding (self): Pfizer; Speaker Bureau/Expert testimony, Research grant/Funding (self): Novartis; Research grant/Funding (self): Roche; Advisory/Consultancy, Research grant/Funding (self): Servier. A.A.M. Van der Veldt: Advisory/Consultancy: BMS; Advisory/Consultancy: MSD; Advisory/Consultancy: Eisai; Advisory/Consultancy: Ipsen; Advisory/Consultancy: Pfizer; Advisory/Consultancy: Roche; Advisory/Consultancy: Novartis; Advisory/Consultancy: Sanofi; Advisory/Consultancy: Pierre Fabre; Advisory/Consultancy: Merck. All other 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.