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

Poster session 08

138P - Toward predicting immune checkpoint blockade response in oesophageal squamous cell carcinoma: Integrating tumour and blood characteristics

Date

14 Sep 2024

Session

Poster session 08

Topics

Cancer Biology;  Tumour Immunology;  Translational Research;  Immunotherapy;  Cancer Diagnostics

Tumour Site

Oesophageal Cancer

Presenters

Amelie Franken

Citation

Annals of Oncology (2024) 35 (suppl_2): S238-S308. 10.1016/annonc/annonc1576

Authors

A. Franken1, N. Meindl-Beinker2, N. Härtel3, M. Ebert2, D. Lambrechts1

Author affiliations

  • 1 Laboratory Of Translational Genetics, VIB-KULeuven Center for Cancer Biology (CCB), 3000 - Leuven/BE
  • 2 Department Of Medicine Ii, Heidelberg University - Faculty of Medicine in Mannheim, 68167 - Mannheim/DE
  • 3 Department Of Medicine Ii, Universitätsklinikum Mannheim, 68167 Mannhe - Mannheim/DE

Resources

Login to get immediate access to this content.

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

Abstract 138P

Background

Immune checkpoint blockade (ICB) holds promise for improving survival rates in oesophageal squamous cell carcinoma (ESCC). However, the lack of robust predictive biomarkers limits clinical outcome optimization. We hypothesized that analysing the spatial organization of tumours can offer insights into ICB response, potentially guiding the identification of reliable non-invasive predictive biomarkers in peripheral blood samples.

Methods

In the RAMONA trial (NCT03416244), tumour sections and peripheral blood samples were collected from 21 ESCC patients before their second-line treatment, either with nivolumab (anti-PD-1) alone or combined with ipilimumab (anti-CTLA4). Tumour sections were stained with the PhenoCycler platform using a 40-plex immunofluorescence panel to examine spatial tumour-immune interactions associated with clinical outcomes, including progression-free survival (PFS) and best overall response by RECIST version 1.1. Peripheral blood mononuclear cells (PBMCs) were isolated and profiled by 10x single-cell RNA-sequencing to compare cellular (sub)types between ICB responders and non-responders.

Results

Patients with prolonged PFS upon ICB (>8 months, n=3) exhibited more intratumoural aggregates of CD3+ T cells (including CD4+ and CD8+ populations), CD79a+ CD38+ B cells, and CD31/CD34+ endothelial cells, compared to patients with shorter PFS (<4 months, n=6; p<0.05). Conversely, tumours from the latter group showed more aggregates of MPO+ neutrophils, in close vicinity to CD4+ FOXP3+ regulatory T cells (p<0.05). These findings were consistent in pre-treatment peripheral blood samples. Specifically, T cells were more abundant in blood samples from patients who experienced prolonged PFS (n=6; p<0.05), while a higher abundance of myeloid cells was associated with shorter PFS (n=9; p<0.05). T cell subclustering revealed that predominantly effector CD8+ T cells and naïve CD4+ T cells correlated with prolonged PFS.

Conclusions

Our study suggests that integrating analyses of both tumour and peripheral blood characteristics can facilitate the discovery of robust non-invasive biomarkers for predicting ICB response.

Clinical trial identification

NCT03416244 (study completion: 2021-11-19).

Editorial acknowledgement

Legal entity responsible for the study

M. Ebert and D. Lambrechts.

Funding

Bristol Myers Squibb.

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.