Impact of NK Cell Activating Receptor Gene Variants on Receptor Expression and Outcome of Immunotherapy in Acute Myeloid Leukemia

Brwa Ali Hussein, Alexander Hallner, Lovisa Wennström, Mats Brune, Anna Martner, Kristoffer Hellstrand, Elin Bernson, Fredrik B Thorén, Brwa Ali Hussein, Alexander Hallner, Lovisa Wennström, Mats Brune, Anna Martner, Kristoffer Hellstrand, Elin Bernson, Fredrik B Thorén

Abstract

Natural killer cells are important effector cells in the immune response against myeloid malignancies. Previous studies show that the expression of activating NK cell receptors is pivotal for efficient recognition of blasts from patients with acute myeloid leukemia (AML) and that high expression levels impact favorably on patient survival. This study investigated the potential impact of activating receptor gene variants on NK cell receptor expression and survival in a cohort of AML patients receiving relapse-preventive immunotherapy with histamine dihydrochloride and low-dose IL-2 (HDC/IL-2). Patients harboring the G allele of rs1049174 in the KLRK1 gene encoding NKG2D showed high expression of NKG2D by CD56bright NK cells and a favorable clinical outcome in terms of overall survival. For DNAM-1, high therapy-induced receptor expression entailed improved survival, while patients with high DNAM-1 expression before immunotherapy associated with unfavorable clinical outcome. The previously reported SNPs in NCR3 encoding NKp30, which purportedly influence mRNA splicing into isoforms with discrete functions, did not affect outcome in this study. Our results imply that variations in genes encoding activating NK cell receptors determine receptor expression and clinical outcome in AML immunotherapy.

Trial registration: ClinicalTrials.gov NCT01347996.

Keywords: Histamine/IL-2; NK cell receptors; Re:Mission trial; acute myeloid leukemia; gene variants; immunotherapy; single nucleotide polymorphism.

Conflict of interest statement

Authors AM, KH, and FT are authors of issued or pending patents protecting the use of histamine dihydrochloride in cancer immunotherapy. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Hussein, Hallner, Wennström, Brune, Martner, Hellstrand, Bernson and Thorén.

Figures

Figure 1
Figure 1
Impact of NKG2D rs1049174 gene variants on outcome of immunotherapy in AML and expression of NKG2D. (A, B) Leukemia-free survival (LFS) and overall survival (OS) of AML patients harboring NKG2D G/x (n=48) or CC (n=32) after receiving HDC/IL-2 treatment. (C–F) NKG2D expression according to NKG2D rs1049174 genotypes in AML patients in both CD16- CD56bright and CD16+CD56+ NK cells before and after immunotherapy as indicated in figures. Patients in figures (C–F) are divided according to indicated number of NKG2D rs1049174 C alleles [n equals 7, 23 and 19 before immunotherapy; panels (C, E); and 8, 23 and 20 after immunotherapy; panels (D, F)]. P values were obtained using log rank test for survival in figures (A, B) and simple linear regression analysis was performed in figures (C–F).
Figure 2
Figure 2
Association between DNAM-1 expression and outcome of immunotherapy in AML. (A, B) Leukemia-free survival (LFS) and overall survival (OS) of AML patients before immunotherapy according to DNAM-1 expression (MFI) in specified NK cell subsets. (C) Box plot shows effect of HDC/IL-2 therapy on median fluorescence intensity (MFI) of DNAM-1 expression in both CD16- CD56bright and CD16+ CD56+ NK cells of AML patients both at the beginning (n=51) and after (n=59) of treatment. (D, E) Impact of induction of the CD16+ CD56+ NK DNAM-1 expression on LFS and OS during first three weeks of immunotherapy in AML. Patients were dichotomized based on above (grey) and below (black) median expression (MFI) of DNAM-1. (F–I) Expression of DNAM-1 in AML patients during immunotherapy based on number of DNAM-1 rs763361 T alleles in CD16-CD56bright and CD16+ CD56+ NK cells before and after receiving one cycle of HDC/IL-2 therapy. Patients are divided according to indicated number of T alleles [n equals 11, 23 and 21 before immunotherapy; panels (F, H); and 12, 24 and 22 after one cycle of immunotherapy; panels (G, I)]. Paired two-sided t test was performed to analyze the difference in DNAM-1 expression before and after therapy in different NK cell subsets in figure (C). Logrank test was carried out for survival analysis in figures (A, B, D, E). Simple linear regression was implemented to investigate impact of DNAM-1 rs763361 gene variants on expression of DNAM-1 both before and after immunotherapy in various NK subsets as shown in figures (F–I).
Figure 3
Figure 3
Impact of NKp30 rs986475 gene polymorphism on expression of NKp30 and clinical outcome of AML during immunotherapy. (A–D) Median fluorescence intensity of NKp30 based on genotype status of NKp30 rs986475 in AML patients in both CD16- CD56bright and CD16+ CD56+ NK cells before and after receiving one cycle of HDC/IL-2 therapy. Patients are divided according to presence (14 out of 61 before therapy, and 15 out of 62 after therapy) or absence of G allele in figures (A–D). (E, F) Kaplan-Meier curves show impact of different NKp30 rs986475 genotypes on leukemia-free survival (LFS) and overall survival (OS) of AML patients after receiving HDC/IL-2 therapy. Simple linear regression was applied to investigate impact of NKp30 gene variants on NKp30 expression both before and after immunotherapy in various NK subsets as shown in figures (A–D). Logrank test was used to analyze the survival based on NKp30 gene variants in figures (E, F).

References

    1. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2015. CA Cancer J Clin (2015) 65(1):5–29. doi: 10.3322/caac.21254
    1. Tallman MS, Wang ES, Altman JK, Appelbaum FR, Bhatt VR, Bixby D, et al. . Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw (2019) 17(6):721–49. doi: 10.6004/jnccn.2019.0028
    1. Breems DA, Van Putten WL, Huijgens PC, Ossenkoppele GJ, Verhoef GE, Verdonck LF, et al. . Prognostic Index for Adult Patients With Acute Myeloid Leukemia in First Relapse. J Clin Oncol (2005) 23(9):1969–78. doi: 10.1200/jco.2005.06.027
    1. Burnett AK, Goldstone A, Hills RK, Milligan D, Prentice A, Yin J, et al. . Curability of Patients With Acute Myeloid Leukemia Who Did Not Undergo Transplantation in First Remission. J Clin Oncol (2013) 31(10):1293–301. doi: 10.1200/jco.2011.40.5977
    1. Wei AH, Döhner H, Pocock C, Montesinos P, Afanasyev B, Dombret H, et al. . Oral Azacitidine Maintenance Therapy for Acute Myeloid Leukemia in First Remission. N Engl J Med (2020) 383(26):2526–37. doi: 10.1056/NEJMoa2004444
    1. DiNardo CD, Wei AH. How I Treat Acute Myeloid Leukemia in the Era of New Drugs. Blood (2020) 135(2):85–96. doi: 10.1182/blood.2019001239
    1. Yu J, Jiang PYZ, Sun H, Zhang X, Jiang Z, Li Y, et al. . Advances in Targeted Therapy for Acute Myeloid Leukemia. Biomark Res (2020) 8. doi: 10.1186/s40364-020-00196-2
    1. Brune M, Castaigne S, Catalano J, Gehlsen K, Ho AD, Hofmann WK, et al. . Improved Leukemia-Free Survival After Postconsolidation Immunotherapy With Histamine Dihydrochloride and Interleukin-2 in Acute Myeloid Leukemia: Results of a Randomized Phase 3 Trial. Blood (2006) 108(1):88–96. doi: 10.1182/blood-2005-10-4073
    1. Martner A, Thorén FB, Aurelius J, Hellstrand K. Immunotherapeutic Strategies for Relapse Control in Acute Myeloid Leukemia. Blood Rev (2013) 27(5):209–16. doi: 10.1016/j.blre.2013.06.006
    1. Aurelius J, Thorén FB, Akhiani AA, Brune M, Palmqvist L, Hansson M, et al. . Monocytic AML Cells Inactivate Antileukemic Lymphocytes: Role of NADPH Oxidase/Gp91(Phox) Expression and the PARP-1/PAR Pathway of Apoptosis. Blood (2012) 119(24):5832–7. doi: 10.1182/blood-2011-11-391722
    1. Aurelius J, Martner A, Brune M, Palmqvist L, Hansson M, Hellstrand K, et al. . Remission Maintenance in Acute Myeloid Leukemia: Impact of Functional Histamine H2 Receptors Expressed by Leukemic Cells. Haematologica (2012) 97(12):1904–8. doi: 10.3324/haematol.2012.066399
    1. Nilsson MS, Hallner A, Brune M, Nilsson S, Thorén FB, Martner A, et al. . Complete Remission After the First Cycle of Induction Chemotherapy Determines the Clinical Efficacy of Relapse-Preventive Immunotherapy in Acute Myeloid Leukaemia. Br J Haematol (2020) 188(4):e49–53. doi: 10.1111/bjh.16320
    1. Ljunggren HG, Kärre K. In Search of the ‘Missing Self’: MHC Molecules and NK Cell Recognition. Immunol Today (1990) 11(7):237–44. doi: 10.1016/0167-5699(90)90097-s
    1. Koch J, Steinle A, Watzi C, Mandelboim O. Activating Natural Cytotoxicity Receptors of Natural Killer Cells in Cancer and Infection. Trends Immunol (2013) 34(4):182–91. doi: 10.1016/j.it.2013.01.003
    1. Hudspeth K, Silva-Santos B, Mavilio D. Natural Cytotoxicity Receptors: Broader Expression Patterns and Functions in Innate and Adaptive Immune Cells. Front Immunol (2013) 4:69. doi: 10.3389/fimmu.2013.00069
    1. Martner A, Rydström A, Riise RE, Aurelius J, Anderson H, Brune M, et al. . Role of Natural Killer Cell Subsets and Natural Cytotoxicity Receptors for the Outcome of Immunotherapy in Acute Myeloid Leukemia. Oncoimmunology (2016) 5(1):e1041701. doi: 10.1080/2162402x.2015.1041701
    1. Fauriat C, Just-Landi S, Mallet F, Arnoulet C, Sainty D, Olive D, et al. . Deficient Expression of NCR in NK Cells From Acute Myeloid Leukemia: Evolution During Leukemia Treatment and Impact of Leukemia Cells in Ncrdull Phenotype Induction. Blood (2007) 109(1):323–30. doi: 10.1182/blood-2005-08-027979
    1. Romero AI, Thorén FB, Brune M, Hellstrand K. Nkp46 and NKG2D Receptor Expression in NK Cells With CD56dim and CD56bright Phenotype: Regulation by Histamine and Reactive Oxygen Species. Br J Haematol (2006) 132(1):91–8. doi: 10.1111/j.1365-2141.2005.05842.x
    1. Crane CA, Han SJ, Barry JJ, Ahn BJ, Lanier LL, Parsa AT, et al. . TGF-Beta Downregulates the Activating Receptor NKG2D on NK Cells and CD8+ T Cells in Glioma Patients. Neuro Oncol (2010) 12(1):7–13. doi: 10.1093/neuonc/nop009
    1. Labani-Motlagh A, Israelsson P, Ottander U, Lundin E, Nagaev I, Nagaeva O, et al. . Differential Expression of Ligands for NKG2D and DNAM-1 Receptors by Epithelial Ovarian Cancer-Derived Exosomes and its Influence on NK Cell Cytotoxicity. Tumour Biol (2016) 37(4):5455–66. doi: 10.1007/s13277-015-4313-2
    1. Mincheva-Nilsson L, Baranov V. Cancer Exosomes and NKG2D Receptor-Ligand Interactions: Impairing NKG2D-Mediated Cytotoxicity and Anti-Tumour Immune Surveillance. Semin Cancer Biol (2014) 28:24–30. doi: 10.1016/j.semcancer.2014.02.010
    1. Deng W, Gowen BG, Zhang L, Wang L, Lau S, Iannello A, et al. . Antitumor Immunity. A Shed NKG2D Ligand That Promotes Natural Killer Cell Activation and Tumor Rejection. Science (2015) 348(6230):136–9. doi: 10.1126/science.1258867
    1. Hayashi T, Imai K, Morishita Y, Hayashi I, Kusunoki Y, Nakachi K, et al. . Identification of the NKG2D Haplotypes Associated With Natural Cytotoxic Activity of Peripheral Blood Lymphocytes and Cancer Immunosurveillance. Cancer Res (2006) 66(1):563–70. doi: 10.1158/0008-5472.Can-05-2776
    1. Espinoza JL, Nguyen VH, Ichimura H, Pham TT, Nguyen CH, Pham TV, et al. . A Functional Polymorphism in the NKG2D Gene Modulates NK-Cell Cytotoxicity and Is Associated With Susceptibility to Human Papilloma Virus-Related Cancers. Sci Rep (2016) 6:39231. doi: 10.1038/srep39231
    1. Löfgren SE, Delgado-Vega AM, Gallant CJ, Sánchez E, Frostegård J, Truedsson L, et al. . A 3’-Untranslated Region Variant Is Associated With Impaired Expression of CD226 in T and Natural Killer T Cells and Is Associated With Susceptibility to Systemic Lupus Erythematosus. Arthritis Rheum (2010) 62(11):3404–14. doi: 10.1002/art.27677
    1. Hafler JP, Maier LM, Cooper JD, Plagnol V, Hinks A, Simmonds MJ, et al. . CD226 Gly307Ser Association With Multiple Autoimmune Diseases. Genes Immun (2009) 10(1):5–10. doi: 10.1038/gene.2008.82
    1. Qiu ZX, Peng Y, Li WM. CD226 Gene Polymorphisms Are Associated With Non-Small-Cell Lung Cancer in the Chinese Han Population. Ther Clin Risk Manag (2015) 11:1259–64. doi: 10.2147/tcrm.S90365
    1. Sanchez-Correa B, Gayoso I, Bergua JM, Casado JG, Morgado S, Solana R, et al. . Decreased Expression of DNAM-1 on NK Cells From Acute Myeloid Leukemia Patients. Immunol Cell Biol (2012) 90(1):109–15. doi: 10.1038/icb.2011.15
    1. Delahaye NF, Rusakiewicz S, Martins I, Ménard C, Roux S, Lyonnet L, et al. . Alternatively Spliced Nkp30 Isoforms Affect the Prognosis of Gastrointestinal Stromal Tumors. Nat Med (2011) 17(6):700–7. doi: 10.1038/nm.2366
    1. Messaoudene M, Fregni G, Enot D, Jacquelot N, Neves E, Germaud N, et al. . Nkp30 Isoforms and Nkp46 Transcripts in Metastatic Melanoma Patients: Unique Nkp30 Pattern in Rare Melanoma Patients With Favorable Evolution. Oncoimmunology (2016) 5(12):e1154251. doi: 10.1080/2162402x.2016.1154251
    1. Aurelius J, Möllgård L, Kiffin R, Ewald F, Sander S, Nilsson FB, et al. . Anthracycline-Based Consolidation may Determine Outcome of Post-Consolidation Immunotherapy in AML. Leuk Lymphoma (2019) 60(11):2771–8. doi: 10.1080/10428194.2019.1599110
    1. Bernson E, Hallner A, Sander FE, Wilsson O, Werlenius O, Rydström A, et al. . Impact of Killer-Immunoglobulin-Like Receptor and Human Leukocyte Antigen Genotypes on the Efficacy of Immunotherapy in Acute Myeloid Leukemia. Leukemia (2017) 31(12):2552–9. doi: 10.1038/leu.2017.151
    1. Bernson E, Hallner A, Sander FE, Nicklasson M, Nilsson MS, Christenson K, et al. . Cytomegalovirus Serostatus Affects Autoreactive NK Cells and Outcomes of IL2-Based Immunotherapy in Acute Myeloid Leukemia. Cancer Immunol Res (2018) 6(9):1110–9. doi: 10.1158/2326-6066.Cir-17-0711
    1. Hallner A, Bernson E, Hussein BA, Ewald F, Sander M, Brune J, et al. . The HLA-B -21 Dimorphism Impacts on NK Cell Education and Clinical Outcome of Immunotherapy in Acute Myeloid Leukemia. Blood (2019) 133(13):1479–88. doi: 10.1182/blood-2018-09-874990
    1. Martner A, Rydström A, Riise RE, Aurelius J, Brune M, Foà R, et al. . NK Cell Expression of Natural Cytotoxicity Receptors may Determine Relapse Risk in Older AML Patients Undergoing Immunotherapy for Remission Maintenance. Oncotarget (2015) 6(40):42569–74. doi: 10.18632/oncotarget.5559
    1. Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural Cytotoxic Activity of Peripheral-Blood Lymphocytes and Cancer Incidence: An 11-Year Follow-Up Study of a General Population. Lancet (2000) 356(9244):1795–9. doi: 10.1016/s0140-6736(00)03231-1
    1. Espinoza JL, Takami A, Onizuka M, Sao H, Akiyama H, Miyamura K, et al. . NKG2D Gene Polymorphism has a Significant Impact on Transplant Outcomes After HLA-Fully-Matched Unrelated Bone Marrow Transplantation for Standard Risk Hematologic Malignancies. Haematologica (2009) 94(10):1427–34. doi: 10.3324/haematol.2009.008318
    1. Carlsten M, Baumann BC, Simonsson M, Jädersten M, Forsblom AM, Hammarstedt C, et al. . Reduced DNAM-1 Expression on Bone Marrow NK Cells Associated With Impaired Killing of CD34+ Blasts in Myelodysplastic Syndrome. Leukemia (2010) 24(9):1607–16. doi: 10.1038/leu.2010.149
    1. Zhou XM, Li WQ, Wu YH, Han L, Cao XG, Yang XM, et al. . Intrinsic Expression of Immune Checkpoint Molecule TIGIT Could Help Tumor Growth In Vivo by Suppressing the Function of NK and CD8(+) T Cells. Front Immunol (2018) 9:2821. doi: 10.3389/fimmu.2018.02821
    1. Sun H, Huang Q, Huang M, Wen H, Lin R, Zheng M, et al. . Human CD96 Correlates to Natural Killer Cell Exhaustion and Predicts the Prognosis of Human Hepatocellular Carcinoma. Hepatology (2019) 70(1):168–83. doi: 10.1002/hep.30347
    1. Enqvist M, Ask EH, Forslund E, Carlsten M, Abrahamsen G, Béziat V, et al. . Coordinated Expression of DNAM-1 and LFA-1 in Educated NK Cells. J Immunol (2015) 194(9):4518–27. doi: 10.4049/jimmunol.1401972
    1. Wagner AK, Kadri N, Snäll J, Brodin P, Gilfillan S, Colonna M, et al. . Expression of CD226 Is Associated to But Not Required for NK Cell Education. Nat Commun (2017) 8:15627. doi: 10.1038/ncomms15627

Source: PubMed

3
Prenumerera