1. Academic Validation
  2. STAT5 promotes PD-L1 expression by facilitating histone lactylation to drive immunosuppression in acute myeloid leukemia

STAT5 promotes PD-L1 expression by facilitating histone lactylation to drive immunosuppression in acute myeloid leukemia

  • Signal Transduct Target Ther. 2023 Sep 30;8(1):391. doi: 10.1038/s41392-023-01605-2.
Ze-Wei Huang 1 Xue-Ning Zhang 1 Ling Zhang 1 Ling-Ling Liu 1 Jing-Wen Zhang 1 Yu-Xiang Sun 2 Jue-Qiong Xu 1 Quentin Liu 3 4 Zi-Jie Long 5
Affiliations

Affiliations

  • 1 Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China.
  • 2 Department of Nephrology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
  • 3 Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China. liuq9@mail.sysu.edu.cn.
  • 4 Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China. liuq9@mail.sysu.edu.cn.
  • 5 Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University; Institute of Hematology, Sun Yat-sen University, Guangzhou, China. longzij@mail.sysu.edu.cn.
Abstract

Immunotherapy is a revolutionized therapeutic strategy for tumor treatment attributing to the rapid development of genomics and immunology, and immune checkpoint inhibitors have successfully achieved responses in numbers of tumor types, including hematopoietic malignancy. However, acute myeloid leukemia (AML) is a heterogeneous disease and there is still a lack of systematic demonstration to apply immunotherapy in AML based on PD-1/PD-L1 blockage. Thus, the identification of molecules that drive tumor immunosuppression and stratify patients according to the benefit from immune checkpoint inhibitors is urgently needed. Here, we reported that STAT5 was highly expressed in the AML cohort and activated the promoter of glycolytic genes to promote glycolysis in AML cells. As a result, the increased-lactate accumulation promoted E3BP nuclear translocation and facilitated histone lactylation, ultimately inducing PD-L1 transcription. Immune checkpoint inhibitor could block the interaction of PD-1/PD-L1 and reactive CD8+ T cells in the microenvironment when co-culture with STAT5 constitutively activated AML cells. Clinically, lactate accumulation in bone marrow was positively correlated with STAT5 as well as PD-L1 expression in newly diagnosed AML patients. Therefore, we have illustrated a STAT5-lactate-PD-L1 network in AML progression, which demonstrates that AML patients with STAT5 induced-exuberant glycolysis and lactate accumulation may be benefited from PD-1/PD-L-1-based immunotherapy.

Figures
Products