AccScience Publishing / EJMO / Volume 7 / Issue 4 / DOI: 10.14744/ejmo.2023.35138
RESEARCH ARTICLE

Disturbed Expression of Memory T-Cell Subsets Could Alter the Outcomes in Adult Acute Myeloid Leukemia

Asmaa M. Zahran1 Zeinab Albadry M. Zahran2 Doaa A. Gamal3 Ibrahim Khalid Ibrahim Elsayh4 Sawsan M. Moeen5 Mohammed M. Wahman6 Marwa A. Sabet7 Azza Mostafa Abdelaal8 Safinaz Hussein5 Khaled Saad8 Amal Rayan3
Show Less
1 Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
2 Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
3 Department of Clinical Oncology, Faculty of Medicine, Assiut University, Assiut, Egypt
4 Faculty of Medicine, Assiut University, Assiut, Egypt
5 Department of Internal Medicine, Clinical Hematology Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
6 Department of Clinical Oncology, Faculty of Medicine, South Valley University, Qena, Egypt
7 Department of Microbiology and Immunology, Faculty of Pharmacy, Sphinx University, New-Assiut, Egypt
8 Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
EJMO 2023, 7(4), 362–370; https://doi.org/10.14744/ejmo.2023.35138
Submitted: 14 October 2023 | Accepted: 13 November 2023 | Published: 29 December 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution -Noncommercial 4.0 International License (CC-by the license) ( https://creativecommons.org/licenses/by-nc/4.0/ )
Abstract

Objectives: We compared the percentages of memory T cells in patients with acute myeloid leukemia (AML) to healthy controls and tried to detect any association of these cells to treatment outcome.

Methods: The study involved 34 adult patients with AML and 24 healthy controls. Following the diagnosis of AML, blood samples were collected from patients and controls for flow cytometric detection of CD8+T, CD4+T, TN, TEM, TCM, TEMRA, and TSCM subsets of both CD4+ and CD8+T-cells.

Results: No significant differences in the mean percentages of CD4+T-cell types between AML patients and controls, with the exception of the total percentage of CD4+T-cells which accumulated in controls, furthermore, significant accumulations of CD8+TEMRA, CD8+CD45+RO, and CD8+TEM were detected in patients compared with controls, while CD8+, CD8+TNs, CD8+TSCM, and CD8+TCM accumulated in controls compared to patients, moreover, significant elevations of total CD8+T-cells, CD8+TEMRA, CD8+TSCM, CD8+TCM, and CD8+TEM in patients with remission compared to those without remission, on the contrary, CD4+T memory cells did not show any significant differences.

Conclusion: Our results showed that accumulation of CD8+T memory cells in AML patients, especially those who achieved remission, could enhance the immune response, particularly in those at high risk of relapse after bone marrow transplantation.

Keywords
Acute myeloid leukemia
CD4+T memory cells
CD8+T memory cells
flow cytometry
remission
Conflict of interest
None declared.
References

1. Duan S, Thomas PG. Balancing immune protection and immune pathology by CD8+ T-cell responses to influenza infection. Front Immunol 2016;7:25.
2. Schmidt ME, Varga SM. The CD8 T cell response to respiratory virus infections. Front Immunol 2018;9:678.
3. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, et al. Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med 1997;186:1407–18.
4. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999;401:708–12. 
5. Pangrazzi L, Naismith E, Meryk A, Keller M, Jenewein B, Trieb K, et al. Increased IL-15 production and accumulation of highly differentiated CD8+ effector/memory T cells in the bone marrow of persons with cytomegalovirus. Front Immunol 2017;8:715.
6. Prlic M, Williams MA, Bevan MJ. Requirements for CD8 T-cell priming, memory generation and maintenance. Curr Opin Immunol 2007;19:315–9.
7. Zahran AM, Abdel-Rahim MH, Nasif KA, Hussein S, Hafez R, Ahmad AB, et al. Association of follicular helper T and follicular regulatory T cells with severity and hyperglycemia in hospitalized COVID-19 patients. Virulence 2022;13(1):569-577.
8. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: Function, generation, and maintenance. Annu Rev Immunol 2004;22:745–63. 
9. Lichtenegger FS, Krupka C, Haubner S, Köhnke T, Subklewe M. Recent developments in immunotherapy of acute myeloid leukemia. J Hematol Oncol 2017;10(1):142–9.
10. Yao D, Xu L, Tan J, Zhang Y, Lu S, Li M, et al. Re-balance of memory T cell subsets in peripheral blood from patients with CML after TKI treatment. Oncotarget 2017;8(47):81852–9.
11. Tan J, Chen S, Lu Y, Yao D, Xu L, Zhang 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;29:463–70.
12. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017;129:424–47.
13. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415–21.
14. Austin R, Smyth MJ, Lane SW. Harnessing the immune system in acute myeloid leukaemia. Crit Rev Oncol Hematol 2016;103:62–77.
15. Mussai F, De Santo C, Abu-Dayyeh I, Booth S, Quek L, McEwenSmith RM, et al. Acute myeloid leukemia creates an arginasedependent immunosuppressive microenvironment. Blood 2013;122:749–58.
16. Kolb HJ, Schattenberg A, Goldman JM, Hertenstein B, Jacobsen N, Arcese W, et al. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood 1995;86:2041–50.
17. Salmon H, Idoyaga J, Rahman A, Leboeuf M, Remark R, Jordan S, et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 2016;44:924–38.
18. Reading JL, Gálvez‐Cancino F, Swanton C, Lladser A, Peggs KS, Quezada SA. The function and dysfunction of memory CD 8+ T cells in tumor immunity. Immunological reviews 2018;283:194–212.
19. Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nature medicine 2015;21:688–97.
20. Noviello M, Manfredi F, Ruggiero E, Perini T, Oliveira G, Cortesi F, et al. Bone marrow central memory and memory stem T-cell exhaustion in AML patients relapsing after HSCT. Nature Communications 2019;10:1065.
21. Gálvez-Cancino F, López E, Menares E, Díaz X, Flores C, Cáceres P, et al. Vaccination-induced skin-resident memory CD8+ T cells mediate strong protection against cutaneous melanoma. Oncoimmunology 2018;7:e1442163.
22. Enamorado M, Iborra S, Priego E, Cueto FJ, Quintana JA, Martínez-Cano S, et al. Enhanced anti-tumour immunity requires the interplay between resident and circulating memory CD8+ T cells. Nature communications 2017;8:1–11.
23. Vahidi Y, Bagheri M, Ghaderi A, Faghih Z. CD8-positive memory T cells in tumor-draining lymph nodes of patients with breast cancer. BMC Cancer 2020;20:257. 
24. Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat Med 2005;11:1299–305.
25. Xu L, Zhang Y, Luo G, Li Y. The roles of stem cell memory T cells in hematological malignancies. J Hematol Oncol 2015;8:113.
26. Horowitz M, Schreiber H, Elder A, Heidenreich O, Vormoor J, Toffalori C, et al. Epidemiology and biology of relapse after stem cell transplantation. Bone Marrow Transplantation 2018;53:1379–89.
27. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, et al. A human memory T cell subset with stem cell–like properties. Nature medicine 2011;17:1290–7.
28. Jansen CS, Prokhnevska N, Master VA, Sanda MG, Carlisle JW, Bilen MA, et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 2019;576:465–70.
29. Peranzoni E, Lemoine J, Vimeux L, Feuillet V, Barrin S, Kantari-Mimoun C, et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti–PD-1 treatment. Proceedings of the National Academy of Sciences 2018;115:e4041–50.
30. Savas P, Virassamy B, Ye C, Salim A, Mintoff CP, Caramia F, et al. Single-cell profiling of breast cancer T cells reveals a tissueresident memory subset associated with improved prognosis. Nature medicine 2018;24:986–93.
31. Eroglu Z, Zaretsky JM, Hu-Lieskovan S, Kim DW, Algazi A, Johnson DB, et al. High response rate to PD-1 blockade in des-moplastic melanomas. Nature 2018;553:347–50.
32. Jin Y, Tan A, Feng J, Xu Z, Wang P, Ruan P, et al. Prognostic impact of memory CD8(+) T Cells on immunotherapy in human cancers: A systematic review and meta-analysis. Front Oncol 2021;11:698076.
33. Martin PJ. Reversing CD8+ T-cell exhaustion with DLI. Blood 2014;123:1289–90. 34. Zahran AM, Shibl A, Rayan A, Mohamed MAEH, Osman AMM, Saad K, et al. Increase in polymorphonuclear myeloid-derived suppressor cells and regulatory T-cells in children with B-cell acute lymphoblastic leukemia. Sci Rep. 2021;11(1):15039.
35. Ge P, Wang W, Li L, Zhang G, Gao Z, Tang Z, et al. Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of colorectal cancer. Biomed Pharmacother 2019;118:109228. 
36. Deng L, Lu D, Bai Y, Wang Y, Bu H, Zheng H. Immune profiles of tumor microenvironment and clinical prognosis among women with triple-negative breast cancer. Cancer Epidemiol Biomarkers Prev 2019;28:1977–85.
37. Choi H, Na KJ. Integrative analysis of imaging and transcriptomic data of the immune landscape associated with tumor metabolism in lung adenocarcinoma: Clinical and prognostic implications. Theranostics 2018;8:1956–65.
38. Ning ZK, Hu CG, Huang C, Liu J, Zhou TC, Zong Z. Molecular subtypes and CD4+ memory T cell-based signature associated with clinical outcomes in gastric cancer. Front Oncol 2021;10:626912.
39. Turtle CJ, Berger C, Sommermeyer D, Hanafi LA, Pender B, Robinson EM, et al. Anti-CD19 chimeric antigen receptormodified T cell therapy for B cell non-Hodgkin lymphoma and chronic lymphocytic leukemia: Fludarabine and cyclophosphamide lymphodepletion improves in vivo expansion and persistence of CAR-T cells and clinical outcomes. Blood 2015;126:184.
40. Turtle CJ, Hanafi LA, Berger C, Sommermeyer D, Pender B, Robinson EM, et al. Addition of fludarabine to cyclophosphamide lymphodepletion improves in vivo expansion of CD19 chimeric antigen receptor-modified T cells and clinical outcome in adults with B cell acute lymphoblastic leukemia. Blood 2015;126:3773.

Share
Back to top
Eurasian Journal of Medicine and Oncology, Electronic ISSN: 2587-196X Print ISSN: 2587-2400, Published by AccScience Publishing