Cyclosporin Influences NK Cell Expansion, Activating and Inhibitory Receptor Expression and Cytokine Synthesis.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3868-3868
Author(s):  
Hongbo Wang ◽  
Alisa Lee ◽  
Michael R. Verneris

Abstract Mouse models demonstrate that natural killer (NK) cells play an important role after allogeneic BMT by mediating GVL effects. Human haploidentical transplantation extends these observations since KIR receptor mismatch between donor and recipient is associated with improved DFS for AML patients. In contrast to the above observations, most patients undergoing hematopoietic cell transplantation receive GVHD prophylaxis with immune suppressive drugs, such as cyclosporin A (CsA). Little is known about the effects of CsA on NK cells and to investigate this, mature peripheral blood NK cells were cultured in IL-2 (1,000 U/ml) with either CsA (1 μg/ml or 10 μg/ml) or vehicle (EtOH) for 7 days. Under these conditions, CsA resulted in a significant inhibition in NK cell (CD3−CD56+) expansion (p<0.05). Cell cycle analysis showed that compared to EtOH, more CsA treated cells were in G1, and less cells were in G2-M phase, demonstrating that CsA reduces the number of NK cells in cycle. Since NK cells recognize malignant targets using both activating and inhibitory cell surface receptors, we used FACS to investigate the expression of KIR receptors (CD158a, CD158b and NKB1) and activating receptors (NKG2D, NKp30, NKp44 and NKp46) on cells cultured with and without CsA. CsA induced changes in the intensity of one or more of the above receptors for all donors tested (n=12). When analyzed in aggregate, we found that compared to EtOH control, NK cells cultured in CsA frequently had reduced expression of KIR receptors (66.7% for CD158a, 50% for CD158b and 33.3% for NKB1) and rarely increased KIR expression (0% for CD158a, 16.6% for CD158b and 0% for NKB1). In contrast, when cells were cultured in CsA the change in expression of NK cell activating receptors was more variable since some receptors increased (33.3% for NKG2D, 33.3% for NKp30, 75% for NKp44 and 16.7% for NKp46) while others receptors decreased (25% for NKG2D, 50% for NKp30, 0% for NKp44 and 16.7% for NKp46). Because CsA affected NK cell receptor density, we performed cytotoxicity assays using both NK cell sensitive (K562) and NK cell resistant, LAK sensitive targets (Raji). NK cells cultured with CsA (for 1 week) had a slightly reduced capacity to kill both targets (E:T 5:1, 60.9%, 36.2%, 35.2% for K562 and 72.4%, 53.3%, 40.7% for EtOH, CsA 1μg/ml and 10μg/ml, respectively). Since CsA changed the expression of NK cell inhibitory and activating receptors, we tested whether this drug would influence the expression of other receptors important in NK cell function. To do this, multiplex PCR was used to analyze the expression of the chemokine receptors SDF-1, CCR 1–4 and CXCR 1–5. Relative to a GADPH control, there was no significant change in chemokine receptor expression after culture with CsA. Lastly, we investigated the effect of CsA on NK cell cytokine synthesis and secretion. Fewer IFN-γ secreting NK cells were present after PMA/ionmycin treatment in CsA containing cultures compared to EtOH controls. Using multiplex PCR, we consistently found that CsA treatment lead to either an induction or an increase in IL-5, IL-6, IL-8, IL-13 and TGF-β transcripts. Taken together these results demonstrate that CsA alters NK cells by inhibiting expansion, changing the density of NK cell inhibitory and activating receptors and shifts cytokine synthesis to a Th2 like pattern.

Blood ◽  
2007 ◽  
Vol 109 (9) ◽  
pp. 3767-3775 ◽  
Author(s):  
Laura Chiossone ◽  
Chiara Vitale ◽  
Francesca Cottalasso ◽  
Sara Moretti ◽  
Bruno Azzarone ◽  
...  

Abstract Steroids have been shown to inhibit the function of fresh or IL-2–activated natural killer (NK) cells. Since IL-15 plays a key role in NK-cell development and function, we comparatively analyzed the effects of methylprednisolone on IL-2– or IL-15–cultured NK cells. Methylprednisolone inhibited the surface expression of the major activating receptors NKp30 and NKp44 in both conditions, whereas NK-cell proliferation and survival were sharply impaired only in IL-2–cultured NK cells. Accordingly, methylprednisolone inhibited Tyr phosphorylation of STAT1, STAT3, and STAT5 in IL-2–cultured NK cells but only marginally in IL-15–cultured NK cells, whereas JAK3 was inhibited under both conditions. Also, the NK cytotoxicity was similarly impaired in IL-2– or IL-15–cultured NK cells. This effect strictly correlated with the inhibition of ERK1/2 Tyr phosphorylation, perforin release, and cytotoxicity in a redirected killing assay against the FcRγ+ P815 target cells upon cross-linking of NKp46, NKG2D, or 2B4 receptors. In contrast, in the case of CD16, inhibition of ERK1/2 Tyr phosphorylation, perforin release, and cytotoxicity were not impaired. Our study suggests a different ability of IL-15–cultured NK cells to survive to steroid treatment, thus offering interesting clues for a correct NK-cell cytokine conditioning in adoptive immunotherapy.


Blood ◽  
2005 ◽  
Vol 106 (13) ◽  
pp. 4370-4376 ◽  
Author(s):  
Sarah Cooley ◽  
Valarie McCullar ◽  
Rosanna Wangen ◽  
Tracy L. Bergemann ◽  
Stephen Spellman ◽  
...  

Although unrelated hematopoietic cell transplantation (HCT) is curative for many hematologic malignancies, complications and relapse remain challenging obstacles. Natural killer (NK) cells, which recover quickly after transplantation, produce cytokines and express killer immunoglobulin-like receptors (KIRs) that regulate their cytotoxicity. Some clinical trials based on a KIR ligand mismatch strategy are associated with less relapse and increased survival, but results are mixed. We hypothesized that T cells in the graft may affect NK cell function and KIR expression after unrelated transplantation and that these differences correlate with clinical outcomes. NK cell function was evaluated using 77 paired samples from the National Marrow Donor Program Research Repository. Recipient NK cells at 100 days after both unmanipulated bone marrow (UBM) and T-cell depleted (TCD) transplants were compared with NK cells from their healthy donors. NK cells expressed fewer KIRs and produced more interferon γ (IFN-γ) after UBM compared to TCD transplants. Multivariate models showed that increased NK cell IFN-γ production correlated with more acute graft-versus-host disease (GVHD), and decreased KIR expression correlated with inferior survival. These results support the notion that T cells in the graft affect NK cell reconstitution in vivo. Understanding these mechanisms may result in strategies to improve clinical outcomes from unrelated HCT.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1417-1417
Author(s):  
Michael Boyiadzis ◽  
Jesse Carson ◽  
Kenton Allen ◽  
Sarfraz A. Memon ◽  
Robert A. Dean ◽  
...  

Abstract NK cells express diverse activating and inhibitory receptors, which collectively determine the NK cytotoxic response. Because NK cells can be potent anti-tumor effectors in the post transplant period of minimal residual disease, we investigated the receptor expression on NK cells following non-myeloablative, HLA-matched, allogeneic hematopoietic stem cell transplant (HSCT). We focused on activating receptors, comparing the expression of natural cytotoxicity receptors (NCR) and C-type lectin receptors on circulating NK cells at one, three, six and twelve months following HSCT with that of the donor-derived mobilized stem cell apheresis. During the first post-transplant month, the absolute numbers of NK cells recovered to normal levels in the 14 patients studied, but the subpopulation of CD56++(bright) CD16− NK cells increased disproportionately compared to the more common CD56+(dull) CD16+ NK cells. By six months the proportions of the NK cell subsets normalized to pre-transplant levels. At one month post transplant, the median percentage of NK cells expressing the anti-tumor NCR NKp46 increased from 15 to 64% and that expressing NKp30 increased from 23 to 40% as compared to the donor’s NK cells; expression remained elevated during the first year. Expression of NKG2D, the homodimeric activating C-type lectin receptor, similarly increased post-transplant. CD94 was also upregulated on NK cells, but the activating heterodimeric partner of CD94, NKG2C, did not change significantly. To investigate these shifts in NK populations and receptor expression, we investigated the effect of cytokine milieu post transplant on these shifts in NK populations and receptor expression. We have determined that plasma levels of IL-15, a cytokine critical in NK differentiation and expansion, increase more than 50-fold in these HSCT patients from pretreatment to the day of transplant and decline in the first weeks post transplant, inversely proportional to NK recovery. When NK cells were cultured with rIL-15, we observed a disproportionate expansion of CD56++ NK cells and a rapid up-regulation of the NCR and NKG2D receptors, similar to the changes observed post-transplant. These data suggest that the cytokine milieu of transplant, in particular elevated levels of IL-15, may contribute to anti-tumor efficacy post transplant by affecting the recovery of NK subsets and modulating expression of activating receptors.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1404-1404
Author(s):  
Hongbo Wang ◽  
Alisa Lee ◽  
Valarie McCullar ◽  
Bruce R. Blazar ◽  
Jeffery S. Miller ◽  
...  

Abstract Following allogeneic hematopoietic cell transplantation, NK cells play important roles in hematopoietic cell engraftment, anti-viral responses, graft vs. host disease (GVHD) and graft vs. leukemia (GVL) reactions. Calcineurin inhibitors, such as cyclosporine A (CsA), are frequently administered to prevent or treat GVHD. It is generally considered that these immune suppressive agents inhibit GVL reactions. To date, little is known about the impact of these immune suppressants on NK cell function. To investigate this, NK cells were isolated from normal donors by negative selection and cultured with IL-2 (100 U/ml), IL-15 (10 ng/ml) and either physiological levels of CsA (1μg/ml) or vehicle control. Under these conditions, we consistently found that CsA treated cultures showed a reduction in NK cell expansion at one week (4.88 vs. 1.87 fold expansion, n=10). The phenotype of CsA treated NK cells was markedly different than controls. More specifically, after 7–10 days of culture with CsA there were significantly more CD56brightCD16− cells and significantly less CD56dimCD16+ cells compared to controls (p<0.001 for both). Accordingly, the percentage of KIR receptor (CD158a/h, CD158b/j and CD158e) expressing cells was significantly less in CsA treated cultures. No consistent changes were detected in the expression of NKG2D, NKp30, NKp44, or NKp46 on CsA treated NK cells relative to controls. To further investigate the influence of CsA on NK cell subset expansion, freshly isolated NK cells were stained with CFSE then cultured with CsA or vehicle control for 1 week. Using FACS we monitored CD56dimCD16+ and CD56brightCD16− cell division. There was no difference in the proliferation of CD56brightCD16− cells between the CsA and control treated cultures. In contrast, the CsA treated CD56dimCD16+ cells had fewer cell divisions, demonstrating that CsA selectively inhibits CD56dimCD16+ cell proliferation. These results were confirmed by determining the fold expansion of freshly isolated, FACS sorted CD56dimCD16+ and CD56brightCD16− populations cultured with CsA or vehicle control. To investigate the cytotoxicity of CsA treated NK cells, we performed killing assays using K562 and Raji cells as targets. Surprisingly, we consistently found higher cytotoxicity in CsA treated NK cells compared to controls (K562, p < 0.05 and Raji, p < 0.05). To further evaluate the functional activity of CsA treated NK cells, we investigated the intracellular IFN-γ secretion following IL-12/IL-18 stimulation. In 5 consecutive experiments the percentage of IFN-γ secreting cells was higher in CsA treated NK cells compared to vehicle controls (44% vs. 24%, p<0.05). Lastly, we determined the effect of CsA on NK cell differentiation from progenitor cells (CD34+Lin−CD38−) using an in vitro differentiation system. Briefly, progenitor cells were cultured on a murine feeder cell line (AFT-024) for 42 days in the presence of IL-3, IL-7, IL-15, SCF and FLT3L +/− CsA. We found that CsA treated cultures had less KIR expressing cells compared to controls. Collectively these results show that physiological levels of CsA inhibit CD56dimCD16+ cell growth and result in a population of NK cells that have less KIR receptor expression, higher cytotoxicity and more cytokine secretion. These findings may have important implications for both GVHD and GVL following hematopoietic cell transplantation.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 8089-8089
Author(s):  
Myo Htut ◽  
Ghislaine Gallez-Hawkins ◽  
Joycelynne Palmer ◽  
Xueli Liu ◽  
Ricardo Tomas Spielberger ◽  
...  

8089 Background: Increased immune responses post autologous HCT may be of benefit in long term disease control. Responses may be mediated by NK cell function and possibly other alternate pathways, including costimulatory molecule pathways. This pilot study assesses the expression of inhibitory (PD-1 and CTLA-4) and stimulatory (OX-40, ICOS, 4-1BB, and CD28) molecules on NK cells after auto-HCT in MM patients and evaluates the effect of lenalidomide treatment on these pathways. Methods: 17 patients with MM undergoing HCT, median age 56.7 years (36 – 67), were included in the study. Peripheral blood samples were taken 3 days prior to HCT and 14, 30, 60, 90, 180 days after HCT. At d180 post-HCT, 13/17 patients were receiving lenalidomide with d91 as median start date. NK cells and their costimulatory molecules were evaluated by flowcytometry using 2 six color panels of antibodies. One way ANOVA test and Kruskal-Wallis test (non-parametric) were applied to analyze the data using the Graphpad Software. Results: See table below. NK cell number was highest (median: 26% of total lymphocytes) at d14 (p: < 0.0001) compared to pre and post HCT levels. At d180, TNF-R OX40 expression was significantly increased in ≤PR group (n=5) (median: 9.5% of NK cells) compared to ≥VGPR (n=12) (0.8%; p=0.0084). In addition, NK cell number was higher in the lenalidomide group (n=13) (median: 15.15 % of total lymphocytes) compared to the no lenalidomide group (n=4) (6.74%; p=0.0108) at d180 post HCT. Significantly lower level of CTLA-4 expression was also found in the lenalidomide group (0.33% vs. 2.54%; p=0.0362). Conclusions: We observed NK cell recovery to baseline values at 60 days after HCT. At d180 post-HCT, OX-40 expression in NK cells was higher in ≤PR group than ≥VGPR group. Lenalidomide treatment was associated with higher NK cells number and decreased expression of CTLA-4. This observation could be a possible marker of enhanced host NK cell immune response against MM. Future clinical trials will explore therapies that increase NK cell responses. [Table: see text]


2009 ◽  
Vol 284 (24) ◽  
pp. 16463-16472 ◽  
Author(s):  
Pedro Roda-Navarro ◽  
Hugh T. Reyburn

NKG2D is an important activating receptor for triggering the NK cell cytotoxic activity, although chronic engagement of specific ligands by NKG2D is also known to provoke decreased cell surface expression of the receptor and compromised NK cell function. We have studied the dynamics of surface NKG2D expression and how exposure to the specific ligand major histocompatibility complex class I chain-related molecule B (MICB) affects receptor traffic and fate. While in the NKL cell line and “resting” NK cells NKG2D was found principally at the cell surface, in activated primary NK cells an intracellular pool of receptor could also be found recycling to the plasma membrane. Exposure of NK cells to targets expressing MICB resulted in degradation of ∼50% of total NKG2D protein and lysosomal degradation of the DAP10 adaptor molecule. Consistent with these observations, confocal microscopy experiments demonstrated that DAP10 trafficked to secretory lysosomes in both transfected NKL cells and in activated primary NK cells upon interaction with MICB-expressing target cells. Interestingly, polarization to the synapse of secretory lysosomes containing DAP10 was also observed. The implications of the intracellular traffic of the NKG2D/DAP10 receptor complex for NK cell activation are discussed. We propose that the rapid degradation of NKG2D/DAP10 observed coincident with recruitment of the receptor to the cytotoxic immune synapse may explain the loss of NKG2D receptor expression after chronic exposure to NKG2D ligands.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5250-5250 ◽  
Author(s):  
Bei Jia ◽  
Chenchen Zhao ◽  
David F. Claxton ◽  
W. Christopher Ehmann ◽  
Witold B. Rybka ◽  
...  

Abstract Natural killer (NK) cells are essential innate immune effectors with promising anti-leukemia activity in acute myeloid leukemia (AML). However, clinical success of applying NK cells in AML treatment has not been achieved. A better understanding of the regulatory mechanisms for NK cell function is important to optimize this therapeutic strategy. T cell immunoglobulin and ITIM domain (TIGIT) is a recently identified inhibitory receptor expressed on T cells and NK cells. Multiple studies including ours have demonstrated its suppressive effect in anti-tumor CD8 T cell response. However whether and how TIGIT impacts NK cells in AML is unknown. Here we performed phenotypic and functional studies on NK cells derived from patients with newly diagnosed AML (n=30). Cells collected from healthy individuals (n=18) were used as controls. TIGIT expression and their contributions to NK cell function in AML were assessed. Peripheral blood samples were first examined by flow cytometry for the frequency of NK cells (defined as CD56+CD3-). The percentage of NK cells among peripheral blood mononuclear cells (PBMCs) in AML patients is comparable with that of healthy controls. In contrast, when we performed functional analysis to assess NK cells for cytokine release upon in vitro stimulation with a human leukemia cell line K562, we observed significantly lower intracellular production of IFN-γ in cells from AML patients compared with that of healthy controls. Consistently NK cells from AML patients expressed less Perforin, indicating a compromised killing capacity. We next evaluated the expression of TIGIT on CD56+CD3- NK cells. As some AML blasts and monocytes also express CD56, we performed multichannel flow cytometry and carefully gated out other cell components when assessing TIGIT expression. To our surprise, we observed a significantly lower frequency of TIGIT-expressing NK cells in AML compared with that of healthy controls (36.82 ±4.543% vs. 48.9±3.818%, P=0.0463). This data indicated that low-TIGIT expression associates with impaired NK cell function and AML progression. We further examined the phenotype and functional status of TIGIT+ NK cells. Expression of activating receptors (CD16 and CD160) and inhibiting receptors (KIR and NKG2A) on TIGIT+ vs. TIGIT- NK cells were analyzed. We observed a significant higher expression of CD16 (51.27±9.009% vs. 20.63±5.334%, P=0.0001) and CD160 (39.84±6.447% vs. 21.24±4.287%, P=0.0103) on TIGIT+ NK cells compared with that of TIGIT- NK cells. By contrast, TIGIT+ NK cells expressed lower KIR (24.06±3.796% vs. 43.59±6.96%, P=0.0046) and NKG2A (7.658±1.717% vs. 18.68±4.256%, P=0.0167) than TIGIT- NK cells. Importantly, functional studies demonstrated an elevated expression of Granzyme B and increased cytokine (IFN-γ and TNF-α) production by TIGIT+ NK cells compared with TIGIT- NK cells (IFN-γ, P=0.0283; TNF-α P=0.0347; Granzyme B, P=0.0493). These data suggest that TIGIT expression on NK cells associated with activated and high functional status. Collectively, our study demonstrates that 1) in line with lower capacity to produce IFN-γ, NK cells from AML patients express less frequency of TIGIT compared with healthy individuals; 2) TIGIT+ NK cells from AML patients express high levels of activating receptors and are highly functional manifested by more cytokine production and enhanced expression of Granzyme B compared with TIGIT- NK cells. These results indicate that in AML patient, TIGIT may contribute to the upregulation of NK cell function. This is in contrast to the observations of CD8 T cells in which TIGIT plays a suppressive role. Targeting TIGIT for cancer treatment is currently under active development. Our findings bring a call for caution on the TIGIT-targeted therapeutic strategy in AML as TIGIT might be a double-edged sword in anti-leukemia immune regulation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4122-4122
Author(s):  
Katja Sockel ◽  
Claudia Schönefeldt ◽  
Sieghart Sopper ◽  
Martin Wermke ◽  
Marc Schmitz ◽  
...  

Abstract Abstract 4122 The hypomethylating agent azacytidine (AZA) represents the standard treatment for many high-risk MDS and AML patients. While the clinical efficacy has been confirmed in several studies, the precise molecular mechanism of action has not been fully understood yet. Human NK-cells play an important role in the regulation of immune responses against malignant cells. Their function is controlled by a complex interplay of activating and inhibitory receptors - some of them being regulated by methylation of the respective genes. We, therefore explored, whether AZA modulates in vitro NK-cell function as well as in vivo during minimal-residual disease (MRD)-guided treatment of imminent relapse in MDS and AML patients treated within the prospective RELAZA trial (NCT00422890). Methods: After purifying NK-cells of healthy donors by MACS (magnetic cell sorting), NK-cells were exposed in vitro to different concentrations of AZA (100nM, 1μM, 3μM) with or without IL-2. In parallel, the NK-cell phenotype of patients (n=12) with AML or MDS, undergoing MRD-guided treatment with AZA after stem cell transplantation was monitored by FACS from peripheral blood samples on day 1, 5 and 7 of the first and second AZA cycle. All patients were still in complete haematological remission at the time of therapy. Results: In vitro, we observed a significant reduction (3,1% to 1,8% p=0.028) of the immature and cytokine-regulating CD56bright NK-cell subpopulation with increasing concentrations of AZA. There was a trend towards a reduced expression of the death-ligand TRAIL, the activating receptors NKG2D and NKp46 and for an increased expression of the inhibitory KIR CD158b1/b2, whereas we could not detect any changes in the expression of FAS-L, Perforin, Granzyme B, NKp30, NKp44, CD69, CD57, DNAM-1, CD16, and NKG2A-CD94. Confirmatory, we observed a significant decrease in the expression of TRAIL (p=0.003), NKG2D (p=0.03) and NKp46 (p=0.006) during AZA treatment in-vivo. Interestingly, these changes appeared to be reversible. The observed reduction of NK-cell activating receptors and TRAIL during AZA treatment correlated with a reduction or stable course of MRD in all analyzed patients. Conclusion: In summary these data suggest that the clinical effects of AZA are not mediated by enhancing NK-cell activity. In fact, the drug may have inhibitory effects on NK-cell function which should be considered when applying AZA in the post-transplant setting. Disclosures: Platzbecker: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


2004 ◽  
Vol 11 (5) ◽  
pp. 879-888 ◽  
Author(s):  
Sunwoong S. Choi ◽  
Vaninder S. Chhabra ◽  
Quoc H. Nguyen ◽  
Bonnie J. Ank ◽  
E. Richard Stiehm ◽  
...  

ABSTRACT Newborn infants have a higher susceptibility to various pathogens due to developmental defects in their host defense system, including deficient natural killer (NK) cell function. In this study, the effects of interleukin-15 (IL-15) on neonatal NK cells was examined for up to 12 weeks in culture. The cytotoxicity of fresh neonatal mononuclear cells (MNC) as assayed by K562 cell killing is initially much less than that of adult MNC but increases more than eightfold after 2 weeks of culture with IL-15 to a level equivalent to that of adult cells. This high level of cytotoxicity was maintained for up to 12 weeks. In antibody-dependent cellular cytotoxicity (ADCC) assays using CEM cells coated with human immunodeficiency virus gp120 antigen, IL-15 greatly increased ADCC lysis by MNC from cord blood. IL-15 increased expression of the CD16+ CD56+ NK markers of cord MNC fivefold after 5 weeks of incubation. Cultures of neonatal MNC with IL-15 for up to 10 weeks resulted in a unique population of CD3− CD8+ CD56+ cells (more than 60%), which are not present in fresh cord MNC. These results show that IL-15 can stimulate neonatal NK cells and sustain their function for several weeks, which has implications for the clinical use of IL-15.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2298-2298
Author(s):  
Miroslaw J. Szczepanski ◽  
Malgorzata Czystowska ◽  
Marta Szajnik ◽  
Ann Welsh ◽  
Kenneth A. Foon ◽  
...  

Abstract Natural killer (NK) cells lyse malignant cells without prior antigen-specific priming and play a critical role in the innate immune response. A balance of signals from activating and inhibiting receptors expressed on each NK cell controls its activity. The growth, differentiation and survival of NK cells have been found to be dependent on interleukin-15 (IL-15). Using multicolor flow cytometry we investigated the receptor repertoire and also measured the NK cell activity in twenty three patients with newly diagnosed acute myeloid leukemia (AML) prior to any treatment. Further, we investigated the ex-vivo effect of IL-15 on the NK cell repertoire and NK cell cytotoxicity. The percentage of circulating NK cells was lower (p&lt;0.0001) in the AML patients (6%± 0.7, range 1–17%) compared to the NK cells of healthy donors (12%± 1, range 9–17%). The expression of the activating natural cytotoxicity (NCR) receptors NKp30 and NKp46 and the C-type lectin receptors NKG2D and NKG2C was significantly decreased in the AML patients compared to the NK cells of healthy donors: NKp30 24 vs 51% p&lt;0.0001, NKp46 32 vs 73% p&lt;0.0001, NKG2D 43 vs 83% p&lt;0.0001, NKG2C 17 vs 28% p&lt;0.03. In addition, the receptor expression (mean fluorescence intensity, MFI) was also significantly lower in AML patients compared to healthy donors. No significant differences in the expression of the NCR NKp44 and the NK- cell inhibitory receptors were observed. Furthermore, the NK cytotoxicity in the AML patients at diagnosis was significantly lower (p&lt;0.0003) compared to the NK cytotoxicity of healthy donors (4 vs 75 LU). When NK cells obtained from AML patients were cultured with IL-15, significant increases in the expression of the NK- cell activating receptors (Table 1) were observed. The upregulation of the activating receptors was associated with a concomitant significant increase (p&lt;0.001) of the NK cell cytotoxicity (4 vs 70 LU). The data suggest that IL-15, a homeostatic NK cell cytokine, can upregulate the expression of activating receptors and concomitantly increase the NK lytic activity. The use of IL-15 as a platform for NK- based therapies for AML patients should be considered in the future. Table 1. The effect of IL-15 on the receptor expression in AML patients NK cell Activating Receptors NK cells in AML pts at diagnosis NK cells in AML pts after IL-15 stimulation p value % positive, MFI % positive, MFI NKp30 24, 2.6 84, 6.0 0.001 NKp44 4, 2.4 71, 6.7 0.001 NKp46 32, 2.6 83, 7 0.002 NKG2C 17, 2.2 58, 6.9 0.005 NKG2D 43, 2.5 87, 7.8 0.01


Sign in / Sign up

Export Citation Format

Share Document