Loss of CD28 expression associates with severe T-cell exhaustion in acute myeloid leukemia

Despite accumulated evidence in T-cell exhaustion in acute myeloid leukemia (AML), the immunotherapeutic targeting exhausted T cells such as programmed cell death protein 1 (PD-1) blockade in AML failed to achieve satisfying efficacy. Characteristics of exhausted T cells in AML remained to be explored.Phenotypic analysis of T cells in bone marrow (BM) using flow cytometry combining senescent and exhausted markers was performed in de novo AML patients and healthy donors as well as AML patients with complete remission (CR). Functional analysis of T-cell subsets was also performed in de novo AML patients using flow cytometry.T cells experienced a phenotypic shift to terminal differentiation characterized by increased loss of CD28 expression and decrease of naïve T cells. Additionally, lack of CD28 expression could help define a severely exhausted subset from generally exhausted T cells (PD-1+TIGIT+). Moreover, CD28- subsets rather than CD28+ subsets predominantly contributed to the significant accumulation of PD-1+TIGIT+ T cells in AML patients. Further comparison of de novo and CR AML patients showed that T-cell exhaustion status was improved after disease remission, especially in CD28+ subsets. Notably, higher frequency of CD28-TIGIT-CD4+ T cells correlated with the presence of minimal residual disease in AML-CR group. However, the correlation between CD28- exhausted T cells and cytogenetic risk or white blood cell count was not observed, except for that CD28- exhausted CD4+ T cells correlated with lymphocyte counts. Intriguingly, larger amount of CD28-TGITI+CD8+ T cells at diagnosis was associated with poor treatment response and shorter leukemia free survival.In summary, lack of CD28 expression defined a severely exhausted status from exhausted T cells. Accumulation of CD28- exhausted T cells was linked to occurrence of AML, and correlated to poor clinical outcome. Our data might facilitate the development of combinatory strategies to improve the efficacy of PD-1 blockade in AML.

[1]  T. Yau,et al.  Immune dysfunction signatures predict outcomes and define checkpoint blockade–unresponsive microenvironments in acute myeloid leukemia , 2022, The Journal of clinical investigation.

[2]  Santiago J. Carmona,et al.  Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. , 2021, Cancer cell.

[3]  S. Salek-Ardakani,et al.  CD8+ T Cell Exhaustion in Cancer , 2021, Frontiers in Immunology.

[4]  P. Fecci,et al.  CD4 T-Cell Exhaustion: Does It Exist and What Are Its Roles in Cancer? , 2021, Clinical Cancer Research.

[5]  G. Kroemer,et al.  Hallmarks of T cell aging , 2021, Nature Immunology.

[6]  C. Bokemeyer,et al.  Increased frequency of TIGIT+CD73-CD8+ T cells with a TOX+ TCF-1low profile in patients with newly diagnosed and relapsed AML , 2021, Oncoimmunology.

[7]  Ying Yue,et al.  Immunosenescence: a key player in cancer development , 2020, Journal of Hematology & Oncology.

[8]  I. Gojo,et al.  Immune escape and immunotherapy of acute myeloid leukemia. , 2020, The Journal of clinical investigation.

[9]  Ling Xu,et al.  T cell senescence and CAR-T cell exhaustion in hematological malignancies , 2018, Journal of Hematology & Oncology.

[10]  Yu Wang,et al.  Reversal of T Cell Exhaustion by the First Donor Lymphocyte Infusion Is Associated with the Persistently Effective Antileukemic Responses in Patients with Relapsed AML after Allo-HSCT. , 2018, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[11]  D. Bigner,et al.  T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in Glioblastoma , 2018, Clinical Cancer Research.

[12]  H. Kantarjian,et al.  Immune therapies in acute myeloid leukemia: a focus on monoclonal antibodies and immune checkpoint inhibitors , 2017, Current opinion in hematology.

[13]  Jessica M. Sido,et al.  CD8+T cells expressing both PD-1 and TIGIT but not CD226 are dysfunctional in acute myeloid leukemia (AML) patients. , 2017, Clinical immunology.

[14]  Shaohua Chen,et al.  Higher PD-1 expression concurrent with exhausted CD8+ T cells in patients with de novo acute myeloid leukemia. , 2017, Chinese journal of cancer research = Chung-kuo yen cheng yen chiu.

[15]  P. Knolle,et al.  TCF1+ hepatitis C virus-specific CD8+ T cells are maintained after cessation of chronic antigen stimulation , 2017, Nature Communications.

[16]  Koichi Araki,et al.  Rescue of exhausted CD8 T cells by PD-1–targeted therapies is CD28-dependent , 2016, Science.

[17]  Bob Löwenberg,et al.  Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. , 2017, Blood.

[18]  R. Scolyer,et al.  Resistance to PD1/PDL1 checkpoint inhibition. , 2017, Cancer treatment reviews.

[19]  R. Greil,et al.  T cells in multiple myeloma display features of exhaustion and senescence at the tumor site , 2016, Journal of Hematology & Oncology.

[20]  J. Wolchok,et al.  Clonal Abundance of Tumor-Specific CD4(+) T Cells Potentiates Efficacy and Alters Susceptibility to Exhaustion. , 2016, Immunity.

[21]  E. Wherry,et al.  Molecular and cellular insights into T cell exhaustion , 2015, Nature Reviews Immunology.

[22]  T. Whiteside,et al.  Programmed death-1 checkpoint blockade in acute myeloid leukemia , 2015, Expert opinion on biological therapy.

[23]  Daniel J Weisdorf,et al.  Acute Myeloid Leukemia. , 2015, The New England journal of medicine.

[24]  S. Henson,et al.  Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? , 2011, Nature Reviews Immunology.