4.7 Article

Control of PD-L1 expression in CLL-cells by stromal triggering of the Notch-c-Myc-EZH2 oncogenic signaling axis

Journal

JOURNAL FOR IMMUNOTHERAPY OF CANCER
Volume 9, Issue 4, Pages -

Publisher

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2020-001889

Keywords

B-lymphocytes; hematologic neoplasms; immunomodulation; programmed cell death 1 receptor; tumor microenvironment

Funding

  1. Gilead research grant
  2. Erich und Gertrud Roggenbuck Stiftung
  3. Deutsche Forschungsgemeinschaft (DFG) [MO 1939/4-1]
  4. Deutsche Krebshilfe [70172788]
  5. Swiss Cancer Research Foundation [KFS-4439-02-2018]
  6. 'Monique-Dornonville de la Cour' Stiftung
  7. CRC
  8. KFSP (Hematology Oncology)
  9. Fondazione AIRC under 5 per Mille 2018 [21198]

Ask authors/readers for more resources

Chronic lymphocytic leukemia (CLL) cells efficiently evade immunosurveillance through close interaction with stromal niches in the bone marrow and lymph nodes. Enhanced PD-L1 expression on CLL cells following stromal contact contributes to immune escape, controlled by a Notch-c-Myc-EZH2 signaling axis. Elevated PD-L1 levels confer resistance towards activated autologous T-cells, highlighting potential therapeutic targets for improving immune responses in CLL patients.
Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults. Emerging data suggest that CLL-cells efficiently evade immunosurveillance. T-cell deficiencies in CLL include immuno(metabolic) exhaustion that is achieved by inhibitory molecules, with programmed cell death 1/programmed cell death ligand 1 (PD-L1) signaling emerging as a major underlying mechanism. Moreover, CLL-cells are characterized by a close and recurrent interaction with their stromal niches in the bone marrow and lymph nodes. Here, they receive nurturing signals within a well-protected environment. We could previously show that the interaction of CLL-cells with stroma leads to c-Myc activation that is followed by metabolic adaptations. Recent data indicate that c-Myc also controls expression of the immune checkpoint molecule PD-L1. Therefore, we sought out to determine the role of stromal contact for the CLL-cells' PD-L1 expression and thus their immuno-evasive phenotype. To do so, we analyzed PD-L1 expression on CLL cell (subsets) in untreated patients and on healthy donor-derived B-cells. Impact of stromal contact on PD-L1 expression on CLL-cells and the underlying signaling pathways were assessed in well-established in vitro niche models. Ex vivo and in vitro findings were validated in the E mu-TCL1 transgenic CLL mouse model. We found increased PD-L1 expression on CLL-cells as compared with B-cells that was further enhanced in a cell-to-cell contact-dependent manner by stromal cells. In fact, circulating recent stromal-niche emigrants displayed higher PD-L1 levels than long-time circulating CLL-cells. Using our in vitro niche model, we show that a novel Notch-c-Myc-enhancer of zeste homolog 2 (EZH2) signaling axis controls PD-L1 upregulation. Ultimately, elevated PD-L1 levels conferred increased resistance towards activated autologous T-cells. In summary, our findings support the notion that the CLL microenvironment contributes to immune escape variants. In addition, several targetable molecules (eg, Notch or EZH2) could be exploited in view of improving immune responses in patients with CLL, which warrants further in-depth investigation.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available