Olink

Olink®
Part of Thermo Fisher Scientific

Immune signatures predict development of autoimmune toxicity in patients with cancer treated with immune checkpoint inhibitors

Med, 2023

Nuñez N., Berner F., Friebel E., Unger S., Wyss N., Gomez J., Purde M., Niederer R., Porsch M., Lichtensteiger C., Kramer R., Erdmann M., Schmitt C., Heinzerling L., Abdou M., Karbach J., Schadendorf D., Zimmer L., Ugurel S., Klümper N., Hölzel M., Power L., Kreutmair S., Capone M., Madonna G., Cevhertas L., Heider A., Amaral T., Hasan Ali O., Bomze D., Dimitriou F., Diem S., Ascierto P., Dummer R., Jäger E., Driessen C., Levesque M., van de Veen W., Joerger M., Früh M., Becher B., Flatz L.

Disease areaApplication areaSample typeProducts
Oncology
Immunotherapy
Patient Stratification
Plasma
Olink Target 96

Olink Target 96

Abstract

Background
Immune checkpoint inhibitors (ICIs) are among the most promising treatment options for melanoma and non-small cell lung cancer (NSCLC). While ICIs can induce effective anti-tumor responses, they may also drive serious immune-related adverse events (irAEs). Identifying biomarkers to predict which patients will suffer from irAEs would enable more accurate clinical risk-benefit analysis for ICI treatment and may also shed light on common or distinct mechanisms underpinning treatment success and irAEs.

Methods
In this prospective multi-center study, we combined a multi-omics approach including unbiased single-cell profiling of over 300 peripheral blood mononuclear cell (PBMC) samples and high-throughput proteomics analysis of over 500 serum samples to characterize the systemic immune compartment of patients with melanoma or NSCLC before and during treatment with ICIs.

Findings
When we combined the parameters obtained from the multi-omics profiling of patient blood and serum, we identified potential predictive biomarkers for ICI-induced irAEs. Specifically, an early increase in CXCL9/CXCL10/CXCL11 and interferon-γ (IFN-γ) 1 to 2 weeks after the start of therapy are likely indicators of heightened risk of developing irAEs. In addition, an early expansion of Ki-67+ regulatory T cells (Tregs) and Ki-67+ CD8+ T cells is also likely to be associated with increased risk of irAEs.

Conclusions
We suggest that the combination of these cellular and proteomic biomarkers may help to predict which patients are likely to benefit most from ICI therapy and those requiring intensive monitoring for irAEs.

Read publication ↗