Article Text
Abstract
Background Our lab and others have shown that a proportion of blood and bone marrow specimens from AML patients have an immunosuppressive microenvironment and have hallmarks of immune exhaustion: increased frequencies of regulatory T cells (Tregs)1 and myeloid-derived suppressor cells (MDSCs),2 decreased T-cell proliferation,3 elevated expression of immune checkpoint molecules4 and increased TEx vs. TPEx5-7 populations. Importantly, a subset of these patient samples containing dysfunctional T cells can be rescued by ICB[3]. We have identified the same immune-related features, in a mouse model of AML. BET inhibitors (BETi) havbeen shown to positively affect CD8+ T cells, indirectly via reduction of PD-L1 expression on myeloid cells8,9 and directly by inhibition of chronic TCR activation genes, increasing stem-cell like memory CD8+ T cells.10,11 We hypothesized that BETi may synergize with anti-PD1 therapy in AML through promoting T cell stemness.
Methods Our AML mouse model bears FLT3-ITD and deletion of TET2 restricted to the myeloid lineage (LysM-CRE). For in vitro studies, splenocytes were stimulated with anti-CD3 and either JQ1, anti-PD1 or both and proliferation and differentiation status were assessed by flow cytometry. For in vivo studies, treatment consisted of 2 weeks. s3-ATAC-seq; Cells were prepared as described.12 Libraries were sequenced on a NextSeq 2000. Data was then analyzed and visualized using the ArchR. GSE Accession: GSE205386.
Results We show that inhibitors which target bromodomain and extra-terminal domain (BET) proteins rescue T cell exhaustion. Ex vivo treatment of cells from AML mice and AML patients with BET inhibitors (BETi) reversed CD8+ T cell exhaustion by restoring proliferative capacity and expansion of the more functional TPEx (figure 1). This reversal is enhanced by combined BETi and anti-PD1 treatment. BETi synergized with anti-PD1 in vivo, resulting in the reduction of circulating leukemia cells, enrichment of CD8+ T cells in the bone marrow, and increased expression of Tcf7, Slamf6, and Cxcr5 in CD8+ T cells (figure 2). Finally, we profiled the epigenomes of in vivo JQ1 treated AML-derived CD8+ T cells by single-cell ATAC seq and find that JQ1 increases Tcf7 accessibility specifically in Tex cells, suggesting that BETi likely mechanistically acts by relieving repression of progenitor programs in Tex CD8+ T cells and maintaining a pool of anti-PD1 responsive CD8 T cells (figure 3).
Conclusions Using an AML mouse model that exhibits leukemia-induced immune exhaustion, we demonstrate the pre-clinical efficacy of combining BETi and anti-PD1 therapy in the treatment of AML.
References
Szczepanski MJ, Szajnik M, Czystowska M, et al. Increased frequency and suppression by regulatory T cells in patients with acute myelogenous leukemia. Clin Cancer Res 2009;15(10):3325–32 doi: 10.1158/1078-0432.CCR-08-3010.
Wang L, Jia B, Claxton DF, et al. VISTA is highly expressed on MDSCs and mediates an inhibition of T cell response in patients with AML. Oncoimmunology 2018;7(9):e1469594 doi: 10.1080/2162402X.2018.1469594.
Lamble AJ, Kosaka Y, Laderas T, et al. Reversible suppression of T cell function in the bone marrow microenvironment of acute myeloid leukemia. Proc Natl Acad Sci U S A 2020;117(25):14331–41 doi: 10.1073/pnas.1916206117.
Williams P, Basu S, Garcia-Manero G, et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2019;125(9):1470–81 doi: 10.1002/cncr.31896.
Noviello M, Manfredi F, Ruggiero E, et al. Bone marrow central memory and memory stem T-cell exhaustion in AML patients relapsing after HSCT. Nat Commun 2019;10(1):1065 doi: 10.1038/s41467-019-08871-1.
Tan J, Chen S, Lu Y, et al. Higher PD-1 expression concurrent with exhausted CD8+ T cells in patients with de novo acute myeloid leukemia. Chin J Cancer Res 2017;29(5):463–70 doi: 10.21147/j.issn.1000-9604.2017.05.11.
Tan J, Yu Z, Huang J, et al. Increased PD-1+Tim-3+ exhausted T cells in bone marrow may influence the clinical outcome of patients with AML. Biomark Res 2020;8:6 doi: 10.1186/s40364-020-0185-8.
Hogg SJ, Vervoort SJ, Deswal S, et al. BET-bromodomain inhibitors engage the host immune system and regulate expression of the immune checkpoint ligand PD-L1. Cell Rep 2017;18(9):2162–74 doi: 10.1016/j.celrep.2017.02.011.
Zhu H, Bengsch F, Svoronos N, et al. BET Bromodomain Inhibition Promotes Anti-tumor Immunity by Suppressing PD-L1 Expression. Cell Rep 2016;16(11):2829–37 doi: 10.1016/j.celrep.2016.08.032.
10. Kagoya Y, Nakatsugawa M, Yamashita Y, et al. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J Clin Invest 2016;126(9):3479–94 doi: 10.1172/JCI86437.
11. Milner JJ, Toma C, Quon S, et al. Bromodomain protein BRD4 directs and sustains CD8 T cell differentiation during infection. J Exp Med 2021;218(8) doi: 10.1084/jem.20202512.
12. Mulqueen RM, Pokholok D, O'Connell BL, et al. High-content single-cell combinatorial indexing. Nat Biotechnol 2021;39(12):1574–80 doi: 10.1038/s41587-021-00962-z.
Ethics Approval IACUC protocol TR01_IP00000907