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

Mini oral session on Developmental and precision medicine

302MO - Development of a flow cytometry-based whole-blood prognostic immune signature in metastatic cancer patients treated with immune checkpoint inhibitors

Date

20 Nov 2020

Session

Mini oral session on Developmental and precision medicine

Topics

Clinical Research;  Targeted Therapy;  Immunotherapy

Tumour Site

Presenters

Jian-Guo Zhou

Citation

Annals of Oncology (2020) 31 (suppl_6): S1358-S1365. 10.1016/annonc/annonc362

Authors

J. Zhou1, A.J. Donaubauer1, B. Frey1, I. Becker1, S. Rutzner1, M. Eckstein1, R. Sun2, H. Ma3, P. Schubert1, C. Schweizer1, R. Fietkau1, E. Deutsch2, U. Gaipl4, M. Hecht1

Author affiliations

  • 1 Department Of Radiation Oncology, Universitätsklinikum Erlangen, 91054 - Erlangen/DE
  • 2 Department Of Radiation Oncology, Institut Gustave Roussy, 94 805 - Villejuif Cedex/FR
  • 3 Department Of Oncology, Zunyi Medical College Affiliated Hospital, 563000 - Zunyi City/CN
  • 4 Department Of Radiation Oncology, Universitätsklinikum Erlangen, 91056 - Erlangen/DE

Resources

Login to get immediate access to this content.

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

Abstract 302MO

Background

The predictive power of programmed cell death ligand 1 (PD-L1) for treatment response to PD-1/PD-L1 immune checkpoint inhibitors (ICI) is not satisfactory. Recent biomarker research focuses on early immunological changes in the peripheral blood to predict treatment response to ICI. Within this prospective ST-ICI trial, pre-planned biomarker analysis was performed and we developed a flow cytometry-based whole-blood prognostic immune signature (FCBPS) to predict overall survival (OS) benefit of cancer patients treated with ICI.

Methods

The predictive power of programmed cell death ligand 1 (PD-L1) for treatment response to PD-1/PD-L1 immune checkpoint inhibitors (ICI) is not satisfactory. Recent biomarker research focuses on early immunological changes in the peripheral blood to predict treatment response to ICI. Within this prospective ST-ICI trial, pre-planned biomarker analysis was performed and we developed a flow cytometry-based whole-blood prognostic immune signature (FCBPS) to predict overall survival (OS) benefit of cancer patients treated with ICI.

Results

A total of 104 patients were prospectively enrolled. Eighty-nine patients provided blood samples. The identified FCBPS signature bases on five immune cell subtypes: neutrophils, plasmacytoid dendritic cells (pDCs), natural killer (NK)T cells (CD56+/CD16+), monocytes (CD14high) and CD8+ T cells (PD-1+). This signature achieved a high accuracy (C-index 0.74 vs 0.71) for predicting OS benefit in the training and validation cohort. Both in the training and validation cohort, the low-risk group had significantly longer OS than the high-risk group (HR 0.26, 95% CI: 0.12-0.56, p=0.00025; HR 0.30, 95% CI: 0.10 -0.91, p=0.024, respectively). In the whole cohort, FCBPS is a predictor of OS (HROS=0.28, 95% CI: 0.15-0.52) and progression-free survival (HRPFS=0.22, 95% CI: 0.12-0.39) that remained independent in multivariate analyses and subgroup analyses after adjusting for clinical and pathological factors.

Conclusions

The flow cytometry-based whole-blood prognostic signature (FCBPS) is a powerful predictor for metastatic cancer patients who benefit from ICI treatment.

Clinical trial identification

NCT03453892; on January 24, 2018.

Editorial acknowledgement

Legal entity responsible for the study

The authors.

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.