-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

2832 Increased Frequency of TOX+ CD39+ TIGIT+ CD73- CD8+ T Cells in Patients with Newly Diagnosed AML

Program: Oral and Poster Abstracts
Session: 613. Acute Myeloid Leukemia: Clinical Studies: Poster III
Hematology Disease Topics & Pathways:
AML, Diseases, immunodeficiency, Biological Processes, Immune Disorders, Technology and Procedures, immune cells, Cell Lineage, Myeloid Malignancies, immune mechanism, flow cytometry, metabolomics, microenvironment
Monday, December 7, 2020, 7:00 AM-3:30 PM

Franziska Brauneck, MD1*, Christin Ackermann, PhD2*, Nils Wildner2*, Jasmin Wellbrock, PhD1*, Carsten Bokemeyer, MD1*, Julian Schulze zur Wiesch, MD2* and Walter Fiedler, MD1

1Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
2Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Immune checkpoint therapy has revolutionized the treatment of patients with cancer. Checkpoint receptors and their ligands play an important role in T cell activation and exhaustion and are currently the focus in understanding the antitumoral immune responses. In patients with acute myeloid leukemia (AML) limited data have been published that comprehensively describe the expression of checkpoint receptors on different T cell subsets.

We performed multicolor flow cytometry on peripheral blood mononuclear cells (PB, PBMCs) from patients with newly diagnosed AML (n=20) and PBMCs from age matched healthy donors (HDs; n=12), focusing on differentiation, the clinically actionable exhaustion receptor T cell immunoglobulin and ITIM domain (TIGIT), the two ectoenzymes ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and ecto-5′-nucleotidase (CD73). Our studies included also analysis of interleukin-7 receptor-α (CD127) and the intracellular expression of the transcription factor T cell factor 1 (TCF-1). Both markers are known to be expressed in long living memory CD8+ T cells and harboring the ability for self-renewal. The thymocyte selection-associated high mobility group box protein (TOX) was also analyzed, since this molecule has recently been described as regulator of CD8+ T cell exhaustion.

Comparison of PB from patients with newly diagnosed AML vs. HDs revealed that the frequency of CD8+, CD4con (CD4+CD127+CD25-) and CD4reg (CD4+CD127-CD25+) T cells was similar among both groups. However, the frequency of CD8+ EMRA T cells (CCR7- CD45RO- CD8+ CD3+) was increased in PB from patients with AML compared to PB from HDs (39,05 ± 4,38 vs. 14,29 ± 3,83; p<0,05).

TIGIT, CD39 and CD73 emerged as checkpoints of interest on CD8+ T cells. The frequency of TIGIT+ CD8+ T cells and CD39+ CD8+ T cells in PBs from patients with newly diagnosed AML was increased compared with that in HDs (42,60 ± 4,67 vs. 20,36 ± 3,68; p=0,00 and 6,54 ± 2,05 vs. 1,31 ± 0,32; p=0,05). Whereas reduced frequency of CD73+ CD8+ T cells occurred in PB from patients with AML vs. HD (41,35 ± 4,75 vs. 66,18 ± 7,28; p=0,01). Analysis between TIGIT and CD73 expression showed inverse correlation between both targets in AML (r=-0,53; p=0,01). The frequency of the TIGIT+ CD73- CD8+ T cell population was increased in AML (36,85 ± 5,17, vs. 16,23 ± 5,09 ;p=0,01). This increased frequency of TIGIT+ CD73- cells in AML was related to EM (CCR7- CD45RO+ CD8+ CD3+)T cells (42,89 ± 4,78 vs. 25,27 ± 3,39; p=0,01) and EMRA CD8+ T cells (56,01 ± 5,68 vs. 36,54 ± 4,85; p=0,03). Moreover, CD39 was aberrantly expressed on this population: we observed an increased frequency of CD39+ TIGIT+ CD73- CD8+ T cells in PB from patients with newly diagnosed AML compared to PB from HDs (13,47 ± 4,03 vs. 3,03 ± 1,21; p=0,02).

Next we focused on CD127 and TCF-1 which are involved in creating long living antigen independent memory CD8+ T cells and ability to self-renewal while producing differentiated effector cells. Comparing expression of CD127 and TCF-1 on CD39+ TIGIT+ CD73- CD8+ T cells showed a significantly decreased frequency of CD127 (17,73 ± 2,16, vs. 30,72 ± 7,12; p=0,04) and TCF-1 (14,67 ± 2,90 vs. 39,74 ± 9,32; p=0,03) in PB from patients with newly diagnosed AML compared to HDs. Expression of TIGIT and TCF-1 inversely correlated in AML (r =-0,87; p<0,05).

To further evaluate the exhaustion status of TIGIT+ CD73- CD8+ T cells we examined the expression of TOX, recently described as one of the key regulators governing CD8+ T cell exhaustion, the frequency of TOX+ cells was increased in AML (50,57 ± 8,21 vs. 22,14 ± 4,86; p=0,01). Analysis of co-expression showed that the TOX+ CD39+ TIGIT+ CD73- CD8+ population was significantly increased in PB from patients with newly diagnosed AML compared to their counterparts in PB from HDs (21,81 ± 3,14 vs. 3,84 ± 1,10; p=0,04).

In summary, we could show that in PB from patients with newly diagnosed AML an aberrant cell population of CD39+ TIGIT+ CD73- CD8+ T cells is prevalent in contrast to the PB from HDs. In this cell population we observed elevated expression of TOX which has recently described as one of the key regulators governing CD8+ T cell exhaustion. In contrast downregulation of CD127 and TCF-1 was found in these cells. These data might contribute to CD8+ T cell exhaustion in AML and support further functional analysis to investigate the relevance of combinatorial inhibition of TIGIT and CD39.

Disclosures: Brauneck: Daiichi Sankyo: Consultancy, Honoraria, Other: support for meeting attendance; Novartis: Other: support for meeting attendance; Jazz Pharmaceuticals: Other: support for meeting attendance. Bokemeyer: Merck KGaA: Honoraria; Janssen-Cilag: Research Funding; Roche: Honoraria, Research Funding; Bayer: Honoraria, Research Funding; Taiho Pharmaceutical: Research Funding; Pfizer: Other; Karyopharm Therapeutics: Research Funding; Millenium: Research Funding; MSD: Research Funding; Nektar: Research Funding; Novartis: Research Funding; Rafael Pharmaceuticals: Research Funding; Springworks Therapeutics: Research Funding; Sanofi: Consultancy, Honoraria, Other: travel accomodations; Bristol-Myers Squibb: Honoraria, Other: travel accomodations, Research Funding; AstraZeneca: Honoraria, Research Funding; Merck Sharp & Dohme: Consultancy, Honoraria; Lilly/ImClone: Consultancy, Research Funding; Merck Serono: Consultancy, Other: travel accomodations; Bayer Schering Pharma: Consultancy; GSO: Consultancy; AOK Health Insurance: Consultancy; Abbvie: Research Funding; ADC Therapeutics: Research Funding; Agile Therapeutics: Research Funding; Alexion Pharmaceuticals: Research Funding; Amgen: Research Funding; Apellis Pharmaceuticals: Research Funding; Astellas: Research Funding; BerGenBio: Research Funding; Blueprint Medicines: Research Funding; Boehringer Ingelheim: Research Funding; Celgene: Research Funding; Daiichi Sankyo: Research Funding; Eisai: Research Funding; Gilead Sciences: Research Funding; Glycotope GmbH: Research Funding; GSK: Research Funding; Incyte: Research Funding; IO Biotech: Research Funding; Isofol Medical: Research Funding. Fiedler: Amgen: Consultancy, Honoraria, Other: support for meeting attendance, Patents & Royalties, Research Funding; ARIAD/Incyte: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Morphosys: Consultancy, Honoraria; Abbvie: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Honoraria, Other: support for meeting attendance; Gilead: Other: support for meeting attendance; Daiichi Sankyo: Other: support for meeting attendance.

<< Previous Abstract | Next Abstract
*signifies non-member of ASH