CD56bright natural killer regulatory cells in filgrastim primed donor blood or marrow products regulate chronic graft- versus-host disease: the Canadian Blood and Marrow Transplant Group randomized 0601 study results

Amina Kariminia, Sabine Ivison, Bernard Ng, Jacob Rozmus, Susanna Sung, Avani Varshney, Mahmoud Aljurf, Sylvie Lachance, Irwin Walker, Cindy Toze, Jeff Lipton, Stephanie J Lee, Jeff Szer, Richard Doocey, Ian Lewis, Clayton Smith, Naeem Chaudhri, Megan K Levings, Raewyn Broady, Gerald Devins, David Szwajcer, Ronan Foley, Sara Mostafavi, Steven Pavletic, Donna A Wall, Stephan Couban, Tony Panzarella, Kirk R Schultz, Amina Kariminia, Sabine Ivison, Bernard Ng, Jacob Rozmus, Susanna Sung, Avani Varshney, Mahmoud Aljurf, Sylvie Lachance, Irwin Walker, Cindy Toze, Jeff Lipton, Stephanie J Lee, Jeff Szer, Richard Doocey, Ian Lewis, Clayton Smith, Naeem Chaudhri, Megan K Levings, Raewyn Broady, Gerald Devins, David Szwajcer, Ronan Foley, Sara Mostafavi, Steven Pavletic, Donna A Wall, Stephan Couban, Tony Panzarella, Kirk R Schultz

Abstract

Randomized trials have conclusively shown higher rates of chronic graft-versus-host disease with filgrastim-stimulated apheresis peripheral blood as a donor source than unstimulated bone marrow. The Canadian Blood and Marrow Transplant Group conducted a phase 3 study of adults who received either filgrastim-stimulated apheresis peripheral blood or filgrastim-stimulated bone marrow from human leukocyte antigen-identical sibling donors. Because all donors received the identical filgrastim dosing schedule, this study allowed for a controlled evaluation of the impact of stem cell source on development of chronic graft-versus-host disease. One hundred and twenty-one evaluable filgrastim-stimulated apheresis peripheral blood and filgrastim-stimulated bone marrow patient donor products were immunologically characterized by flow cytometry and tested for their association with acute and chronic graft-versus-host disease within 2 years of transplantation. The immune populations evaluated included, regulatory T cells, central memory and effector T cells, interferon γ positive producing T cells, invariate natural killer T cells, regulatory natural killer cells, dendritic cell populations, macrophages, and activated B cells and memory B cells. When both filgrastim-stimulated apheresis peripheral blood and filgrastim-stimulated bone marrow were grouped together, a higher chronic graft-versus-host disease frequency was associated with lower proportions of CD56bright natural killer regulatory cells and interferon γ-producing T helper cells in the donor product. Lower CD56bright natural killer regulatory cells displayed differential impacts on the development of extensive chronic graft-versus-host disease between filgrastim-stimulated apheresis peripheral blood and filgrastim-stimulated bone marrow. In summary, while controlling for the potential impact of filgrastim on marrow, our studies demonstrated that CD56bright natural killer regulatory cells had a much stronger impact on filgrastim-stimulated apheresis peripheral blood than on filgrastim-stimulated bone marrow. This supports the conclusion that a lower proportion of CD56bright natural killer regulatory cells results in the high rate of chronic graft-versus-host disease seen in filgrastim-stimulated apheresis peripheral blood. clinicaltrials.gov Identifier: 00438958.

Trial registration: ClinicalTrials.gov NCT00438958.

Copyright© Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
Algorithm of sample analysis for immune population that correlate with the development of overall cGvHD. G-PB: G peripheral blood; cGvHD: chronic graft-versus-host disease; aGvHD: acute graft-versus-host disease; IFN: interferon; NK: natural killer; DC: dendritic cells; TRM: transplant related mortality.
Figure 2.
Figure 2.
Correlation of donor CD56bright NKreg cells infusion characteristics with acute and chronic GvHD. (A) Box and whisker plot of CD56bright NKreg cells percentage per total lymphocytes for overall cGvHD, which was calculated as the total nucleated cells gated on FSC/SSC plots, then the percentage of each subpopulation of interest was determined as a percentage of the total number of lymphocytes (based on forward and side scatter). The phenotype of CD56bright NK cells was CD56bright CD3-CD16-perforin-granzyme B. (B) The correlation of CD56bright NKreg cells with extensive cGvHD. (C) Representative dot plot showing CD56bright and CD56dim subpopulations of NKreg cells. Mononuclear cells were stained by appropriate conjugated mAbs for surface markers. The cells were then fixed and treated to be permeable, followed by intracellular staining (Online Supplementary Table S1). The data was acquired by LSRII equipped with four lasers. A minimum of 1×10 cells were acquired. Single cells were gated based on FSC-A vs. FSC-H and dead cells were excluded using fixable viability dye. Lymphocytes were gated as FSC lo and SSC lo. Upper left dot plot: NK cells were defined as CD3−CD56+ cells. Two populations are revealed based on expression of CD56, CD56bright (gate A) and CD56dim (gate B). CD56brightcells express a significantly higher level of activating receptor CD335 (NKp46) compared to CD56dim. CD56bright cells express a significantly lower level of molecules involved in killing, Granzyme B (bottom left dot plot) and Perforin (bottom right dot plot). (D) A ROC analysis was used to determine an ‘optimal’ cut point for correctly predicting the occurrence of overall chronic GvHD. ‘Optimal’ was defined as the point on the ROC curve with the shortest distance from the point (0, 1). The point (0, 1) represents the ideal, 100% sensitivity and 100% specificity. GvHD: graft-versus-host disease; NK: natural killer; AUC: area under the curve.
Figure 3.
Figure 3.
Correlation of donor IFNγ producing CD4+ T cells infusion characteristics with acute and chronic GvHD. (A) Box and whisker plot of the correlation of IFNγ producing CD4+ T cells percentage with overall cGvHD. The population was calculated as a subpopulation of CD3+ lymphocytes. PBMCs were stimulated with PMA (100ng/ml)/Ionomycin (1 μg/ml) in the presence of monensin for 6 hours. Unstimulated cells treated with monensin were used as control. At the end of incubation, the cells were harvested and surface markers were stained for CD3, CD4, and CD8, in addition to fixable viability to distinguish dead cells. Intracellular staining was performed to detect production of IL-17, IL-4 and IFNγ. The data was acquired using BD LSRII and analyzed by FlowJo v9. Hierarchical gating; 1: lymphocytes were selected based on FSC/SSC, 2: exclusion of dead cells, 3: selection of CD3+ T cells, and 4: determination of IFNγ producing CD4+ T-cells. The data is presented as % of CD4+IFNγ+ T cells per CD3+ lymphocytes. (B) Correlation of IFNγ producing CD4+ T cells with extensive cGvHD; (C) A representative dot plot of INFγ+ T helper cells. Mononuclear cells were seeded at a density of 1×10 per milliliter of culture medium (RPMI-1640 supplemented with 10% heat-inactivated FBS and 2mM l-glutamine), and the cells incubated in CO2 incubator providing 95% oxygen and 5% CO2 at 37 degrees centigrade. The cells were stimulated for 6 hours with 100ng/ml PMA and 1 μg/ml Ionomycin in presence of golgi inhibitor monensin (BD Biosciences; following manufacturers’ instructions). As control, cells were cultured without stimulator, but received monensin. The cells were then harvested and surface staining was performed to detect CD3+CD4+ cells. Then the cells were fixed and treated to be permeable. Intracellular staining was performed to detect IFNγ, IL-4 and IL-17. At least 1×10 cells were acquired. Gating hierarchy; 1: single cells (FSC-A vs. FSC-H), 2: viable cells (fixable viability dye negative), and 3: lymphocytes 4-CD3+CD4+ cells. The percentage of Th1 (IFNγ+), Th17 (IL-17+) and Th2 (IL-4+) were determined after setting quadrant based on unstimulated cells (upper row). Dot plots in lower row show cytokine expression after stimulation. D) A ROC analysis was used to determine an ‘optimal’ cut point for correctly predicting the occurrence of chronic GvHD. The optimal cut point for IFNγ producing CD4+ T cells/CD3+ T cells was 13.9%. (E) Logistic regression was performed with the two identified markers as predictors and overall cGvHD status as response. Each sample was given an estimated probability of overall cGvHD based on the fitted model. The ROC was generated by applying different probability thresholds. GvHD: graft-versus-host disease; IFN: interferon; AUC: area under the curve.
Figure 4.
Figure 4.
Impact of donor IFNγ producing CD4+ T cells and CD56bright NKreg cells infusion characteristics on cGvHD by donor source using G-PB or G-BM transplantation. (A) The estimated probability of overall cGvHD by treatment (donor source G-BM versus G-PB) as a function of the CD56bright cells per total lymphocytes. B) Estimated probability of extensive cGvHD as a function of CD56bright cells per total lymphocytes by donor source (G-PB or G-BM). C) Estimated probability of overall cGvHD as a function of donor IFNγ producing CD4+ T cells by donor source (G-PB or G-BM); D) Estimated probability of extensive cGvHD as a function of donor IFNγ producing CD4+ T cells by donor source (G-PB or G-BM). GvHD: graft-versus-host disease; IFN: interferon.

References

    1. Lee SJ, Logan B, Westervelt P, et al. Patient-reported outcomes in 5-year survivors who received bone marrow vs. peripheral blood unrelated donor transplantation: long-term follow-up of a randomized clinical trial. JAMA Oncol. 2016;2(12):1583–1589.
    1. Burns LJ, Logan BR, Chitphakdithai P, et al. Recovery of unrelated donors of peripheral blood stem cells versus recovery of unrelated donors of bone marrow: a prespecified analysis from the Phase III Blood and Marrow Transplant Clinical Trials Network Protocol 0201. Biol Blood Marrow Transplant. 2016;22(6):1108–1116.
    1. Anasetti C, Logan BR, Lee SJ, et al. Blood and Marrow Transplant Clinical Trials Network. Peripheral-blood stem cells versus bone marrow from unrelated donors. N Engl J Med. 2012;367(16):1487–1496.
    1. Couban S, Simpson DR, Barnett MJ, et al. Canadian Bone Marrow Transplant Group A randomized multicenter comparison of bone marrow and peripheral blood in recipients of matched sibling allogeneic transplants for myeloid malignancies. Blood. 2002;100(5):1525–1531.
    1. Chu R, Brazauskas R, Kan F, et al. Comparison of outcomes after transplantation of G-CSF-stimulated bone marrow grafts versus bone marrow or peripheral blood grafts from HLA-matched sibling donors for patients with severe aplastic anemia. Biol Blood Marrow Transplant. 2011;17(7):1018–1024.
    1. Nagafuji K, Matsuo K, Teshima T, et al. Peripheral blood stem cell versus bone marrow transplantation from HLA-identical sibling donors in patients with leukemia: a propensity score-based comparison from the Japan Society for Hematopoietic Stem Cell Transplantation registry. Int J Hematol. 2010;91(5):855–864.
    1. Eapen M, Logan BR, Confer DL, et al. Peripheral blood grafts from unrelated donors are associated with increased acute and chronic graft-versus-host disease without improved survival. Biol Blood Marrow Transplant. 2007;13(12):1461–1468.
    1. Schrezenmeier H, Passweg JR, Marsh JC, et al. Worse outcome and more chronic GVHD with peripheral blood progenitor cells than bone marrow in HLA-matched sibling donor transplants for young patients with severe acquired aplastic anemia. Blood. 2007;110(4):1397–1400.
    1. Goldman J. Peripheral blood stem cells for allografting. Blood. 1995;85(6):1413–1415.
    1. Dhédin N, Prébet T, De Latour RP, et al. Extensive chronic GVHD is associated with donor blood CD34+ cell count after G-CSF mobilization in non-myeloablative allogeneic PBSC transplantation. Bone Marrow Transplant. 2012;47(12):1564–1568.
    1. Mohty M, Bilger K, Jourdan E, et al. Higher doses of CD34+ peripheral blood stem cells are associated with increased mortality from chronic graft-versus-host disease after allogeneic HLA-identical sibling transplantation. Leukemia. 2003;17(5):869–875.
    1. Vasu S, Geyer S, Bingman A, et al. Granulocyte colony-stimulating factor-mobilized allografts contain activated immune cell subsets associated with risk of acute and chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2016;22(4):658–668.
    1. Gallo S, Woolfrey AE, Burroughs LM, et al. Marrow grafts from HLA-identical siblings for severe aplastic anemia: does limiting the number of transplanted marrow cells reduce the risk of chronic GvHD? Bone Marrow Transplant. 2016;51(12):1573–1578.
    1. Arpinati M, Chirumbolo G, Urbini B, et al. Acute graft-versus-host disease and steroid treatment impair CD11c+ and CD123+ dendritic cell reconstitution after allogeneic peripheral blood stem cell transplantation. Biol Blood Marrow Transplant. 2004;10(2):106–115.
    1. Ding L, Zhu H, Yang Y, et al. The absolute number of regulatory T cells in unmanipulated peripheral blood grafts predicts the occurrence of acute graft-versus-host disease post haplo-identical hematopoietic stem cell transplantation. Leuk Res. 2017;56:13–20.
    1. Vela-Ojeda J, García-Ruiz Esparza MA, Reyes-Maldonado E, et al. Clinical relevance of NK, NKT, and dendritic cell dose in patients receiving G-CSF-mobilized peripheral blood allogeneic stem cell transplantation. Ann Hematol. 2006;85(2):113–120.
    1. Couban S, Aljurf M, Lachance S, et al. Filgrastim-stimulated bone marrow compared with filgrastim-mobilized peripheral blood in myeloablative sibling allografting for patients with hematologic malignancies: a randomized Canadian Blood and Marrow Transplant Group Study. Biol Blood Marrow Transplant. 2016;22(8):1410–1415.
    1. Przepiorka D, Weisdorf D, Martin P, et al. Consensus conference on acute GVHD grading bone marrow transplant. 1995;15:825–828.
    1. Sullivan K. Acute and chronic graft versus host disease in man. Int J Cell Cloning 1986;4 Suppl 1:42–93.
    1. Cox DR, Oakes D. 1984. Analysis of Survival Data. New York: Chapman & Hall; ISBN 041224490X.
    1. Yakoub-Agha I1, Saule P, Depil S, et al. Comparative analysis of naïve and memory CD4+ and CD8+ T-cell subsets in bone marrow and G-CSF-mobilized peripheral blood stem cell allografts: impact of donor characteristics. Exp Hematol. 2007;35(6):861–871.
    1. Shaw BE, Apperley JF, Russell NH, et al. Unrelated donor peripheral blood stem cell transplants incorporating pre-transplant in-vivo alemtuzumab are not associated with any increased risk of significant acute or chronic graft-versus-host disease. Br J Haematol. 2011;153(2):244–252.
    1. Rozmus J, Schultz KR, Wynne K, et al. Early and late overall chronic graft-versus-host disease (overall cGvHD) in children is characterized by different Th1/Th2 cytokine profiles: findings of The Children’s Oncology Group Study (COG), ASCT0031. Biol Blood Marrow Transplant. 2011;17(12):1804–1813.
    1. Bacigalupo A, Lamparelli T, Barisione G, et al. Gruppo Italiano Trapianti Midollo Osseo (GITMO). Thymoglobulin prevents chronic graft-versus-host disease, chronic lung dysfunction, and late transplant-related mortality: long-term follow-up of a randomized trial in patients undergoing unrelated donor transplantation. Biol Blood Marrow Transplant. 2006;12(5):560–565.
    1. Wolschke C, Zabelina T, Ayuk F, et al. Effective prevention of GVHD using in vivo T-cell depletion with anti-lymphocyte globulin in HLA-identical or -mismatched sibling peripheral blood stem cell transplantation. Bone Marrow Transplant. 2014;49(1):126–130.
    1. Devillier R, Granata A, Fürst S, et al. Low incidence of chronic GVHD after HLA-haploidentical peripheral blood stem cell transplantation with post-transplantation cyclophosphamide in older patients. Br J Haematol. 2017;176(1):132–135.
    1. Kanakry CG, O’Donnell PV, Furlong T, et al. Multi-institutional study of post-transplantation cyclophosphamide as single-agent graft-versus-host disease prophylaxis after allogeneic bone marrow transplantation using myeloablative busulfan and fludarabine conditioning. J Clin Oncol. 2014;32(31):3497–3505.
    1. Carnevale-Schianca F, Caravelli D, Gallo S, et al. Post-transplant cyclophosphamide and tacrolimus-mycophenolate mofetil combination prevents graft-versus-host disease in allogeneic peripheral blood hematopoietic cell transplantation from HLA-matched donors. Biol Blood Marrow Transplant. 2017;23(3):459–466.
    1. Bashey A, Zhang X, Sizemore CA, et al. T-cell-replete HLA-haploidentical hematopoietic transplantation for hematologic malignancies using post-transplantation cyclophosphamide results in outcomes equivalent to those of contemporaneous HLA-matched related and unrelated donor transplantation. J Clin Oncol. 2013;31(10):1310–1316.
    1. Li Pira G, Malaspina D, Girolami E, et al. Selective Depletion of αβ T Cells and B Cells for Human Leukocyte Antigen-Haploidentical Hematopoietic Stem Cell Transplantation. A Three-Year Follow-Up of Procedure Efficiency. Biol Blood Marrow Transplant. 2016;22(11):2056–2064.
    1. Arai S, Sahaf B, Narasimhan B, et al. Prophylactic rituximab after allogeneic transplantation decreases B-cell alloimmunity with low chronic GVHD incidence. Blood. 2012;119(25):6145–6154.
    1. Delia M, Pastore D, Mestice A, et al. Outcome of allogeneic peripheral blood stem cell transplantation by donor graft CD3+/Tregs ratio: a single-center experience. Biol Blood Marrow Transplant. 2013;19(3):495–499.
    1. Nachbaur D, Kircher B. Dendritic cells in allogeneic hematopoietic stem cell transplantation. Leuk Lymphoma. 2005;46(10):1387–1396.
    1. She K, Gilman AL, Aslanian S, et al. Altered Toll-like receptor 9 responses in circulating B cells at the onset of pediatric chronic GVHD. Biol Blood Marrow Transplant. 2007;13(4):386–397.
    1. Shier LR, Schultz KR, Imren S, et al. Differential effects of granulocyte colony-stimulating factor on marrow- and blood-derived hematopoietic and immune cell populations in healthy human donors. Biol Blood Marrow Transplant. 2004;10(9):624–634.
    1. Waller EK, Logan BR, Harris WA, et al. Improved survival after transplantation of more donor plasmacytoid dendritic or naïve T cells from unrelated-donor marrow grafts: results from BMTCTN 0201. J Clin Oncol. 2014;32(22):2365–2372.
    1. Baume DM, Robertson MJ, Levine H. Differential responses to interleukin 2 define functionally distinct subsets of human natural killer cells. Eur J Immunol. 1992;22(1):1–6.
    1. Larghero J, Rocha V, Porcher R, et al. Association of bone marrow natural killer cell dose with neutrophil recovery and chronic graft-versus-host disease after HLA identical sibling bone marrow transplants. Br J Haematol. 2007;138(1):101–109.
    1. Karrich JJ, Cupedo T. Group 3 innate lymphoid cells in tissue damage and graft-versus-host disease pathogenesis. Curr Opin Hematol. 2016;23(4):410–415.
    1. Munneke JM, Björklund AT, Mjösberg JM, et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood. 2014;124(5):812–821.
    1. Kariminia A, Holtan SG, Ivison S, et al. Heterogeneity of chronic graft-versus-host disease biomarkers: the only consistent association is with CXCL10 and CXCR3+ NK cells. Blood. 2016;127(24):3082–3089.
    1. Moretta F, Petronelli F, Lucarelli B, et al. The generation of human innate lymphoid cells is influenced by the source of hematopoietic stem cells and by the use of G-CSF. Eur J Immunol. 2016;46(5):1271–1278.
    1. Bogunia-Kubik K, Mlynarczewska A, Wysoczanska B, Lange A. Recipient interferon-gamma 3/3 genotype contributes to the development of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Haematologica. 2005;90(3):425–426.
    1. Baker J, Verneris MR, Ito M, Shizuru JA, Negrin RS. Expansion of cytolytic CD8(1) natural killer T cells with limited capacity for graft- versus-host disease induction due to interferon gamma production. Blood. 2001;97(10):2923–2931.
    1. Lu Y, Waller EK. Dichotomous role of interferon-gamma in allogeneic bone marrow transplant. Biol Blood Marrow Transplant. 2009;15(11):1347–1353.
    1. Fu J, Wang D, Yu Y, et al. T-bet is critical for the development of acute graft-versus-host disease through controlling T cell differentiation and function. J Immunol. 2015;194(1):388–397.
    1. Hall BM, Tran GT, Verma ND, et al. Do natural T regulatory cells become activated to antigen specific T regulatory cells in transplantation and in autoimmunity? Front Immunol. 2013;4:208.
    1. Wang H, Yang YG. The complex and central role of interferon-γ in graft-versus-host disease and graft-versus-tumor activity. Immunol Rev. 2014;258(1):30–44.
    1. Cope A, Le Friec G, Cardone J, Kemper C. The Th1 life cycle: molecular control of IFN-γ to IL-10 switching. Trends Immunol. 2011;32(6):278–286.
    1. Chinnadurai R, Copland IB, Patel SR, Galipeau J. IDO-independent suppression of T-cell effector function by IFN-γ licensed human mesenchymal stromal cells. J Immunol. 2014;192(4):1491–1501.

Source: PubMed

3
S'abonner