Abstract

Natural killer (NK) cells are important for early tumor immune surveillance. In patients with hematological cancers, NK cells are generally functional deficient and display dysregulations in their receptor repertoires. Acute leukemia is the most common cancer in children, and we here performed a comparative phenotypic profiling of NK cells from B-cell precursor acute lymphoblastic leukemia (BCP-ALL) patients to identify aberrant NK cell phenotypes. NK cell phenotypes, maturation, and function were analyzed in matched bone marrow and blood NK cells from BCP-ALL patients at diagnosis, during treatment, and at end of treatment and compared with age-matched pediatric control subjects. Expression of several markers were skewed in patients, but with large interindividual variations. Undertaking a multiparameter approach, we found that high expression levels of NKG2A was the single predominant marker distinguishing NK cells in BCP-ALL patients compared with healthy control subjects. Moreover, naïve CD57NKG2A NK cells dominated in BCP-ALL patients at diagnosis. Further, we found dysregulated expression of the activating receptor DNAM-1 in resident bone marrow CXCR6+ NK cells. CXCR6+ NK cells lacking DNAM-1 expressed NKG2A and had a tendency for lower degranulation activity. In conclusion, high expression of NKG2A dominates NK cell phenotypes from pediatric BCP-ALL patients, indicating that NKG2A could be targeted in therapies for this patient group.

1. Introduction

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Improved risk-adapted treatment protocols aimed at reducing toxicity in low-risk patients while providing more aggressive therapy in high-risk patients have led to 5-year overall survival above 90% for non–high-risk pediatric precursor–B cell acute lymphoblastic leukemia (BCP-ALL).1 Within the BCP-ALL group, around 10% to 15% of children achieving remission will relapse, which may occur despite beneficial scores of residual disease during treatment.

Natural killer (NK) cell functional defects are reported in patients with hematological cancers such as ALL, acute myeloid leukemia (AML) and chronic myeloid leukemia (CML), and myelodysplastic syndrome,2–6 and have been correlated to increased risk of relapse.6,7 NK cells target malignant cells when signals through activating receptors overrides inhibition through inhibitory receptors engaging major histocompatibility complex class I. Low expression of activating receptors on NK cells at diagnosis correlate to the observed low NK cell activity in patients with AML and ALL.2,4–8 In terms of acute leukemia, the activating receptors DNAM-1, NKp46, and NKp30 are suggested to mediate NK cell recognition of blasts. In context of acute leukemia, expression of activating receptors are reduced on NK cells at diagnosis in both adult AML patients and in pediatric BCP-ALL patients, which is linked to ligand-induced downregulation of the receptors.9–14 Low surface expression of activating receptors at diagnosis further correlate to low NK cell activity and increased risk of relapse.4–8 In contrast, the inhibitory NKG2A receptor is expressed at higher levels in NK cells of both adult AML and pediatric BCP-ALL5,15 and is linked to increased risk of remission failure in adult AML patients.5 Understanding the role of endogenous NK cells in controlling leukemia is important to design therapeutic protocols aimed at increasing NK cell–tumor activity. While allogeneic NK cells have been successfully used in therapy, there is also a potential for exploiting the patient's own NK cells to more efficiently target the blasts. We have previously shown that it is possible to sensitize autologous NK cells to resistant ALL blasts.2

There are 2 major populations of circulating NK cells, CD56bright and CD56dim subsets, in which the latter is considered the most differentiated form. Within the CD56dim subset, populations at varying maturational stages exist, in which maturation follows a path of gradual loss of the inhibitory receptor NKG2A and increased expression of CD57 and KIR.16,17 Increased proportions of naïve CD57 NKG2A+KIR NK cells at diagnosis have been correlated to lower overall survival in adult AML patients.18 In contrast, a predominant naïve CD57NKG2A+ phenotype in recipients after hematopoietic stem cell transplantation is associated with increased overall survival.19

Bone marrow (BM) additionally contains a distinct population of resident NK cells that uniquely expresses CD69 and the chemokine receptor CXCR6, both markers absent on circulating NK cells.20 CD69 is important for tissue retention of lymphocytes by preventing sphingosine-1-phosphate receptor–dependent egress into blood.21 CXCR6+CD69+ NK cells are also found in spleen and lymph nodes and resemble tissue resident NK cells recently defined in multiple nonlymphoid tissues such as liver, lung, uterus, and nasal cavity.22–28 How this subset is affected by an acute leukemia is currently unknown.

In this study, we analyzed NK cell phenotypes, maturation markers, and cellular function in pediatric BCP-ALL patients at diagnosis and throughout treatment. Matched BM and blood NK cells were compared with gain insights into putative differences between circulating NK cells and NK cells present in the leukemic niche. We found that high expression levels of NKG2A was the most consistent marker of NK cells at diagnosis. Our study indicates that NKG2A represents a target for therapeutic intervention in pediatric acute leukemia.

2. Methods

2.1 Patients and healthy control subjects

Peripheral venous blood (5 mL) and BM aspirates (1 mL) were collected from pediatric BCP-ALL patients (Table 1) admitted between January 2014 and November 2017 at Oslo University Hospital (Oslo, Norway) during routine diagnostic procedures. Peripheral venous blood (n = 57) and BM aspirates (n = 10) were collected from healthy age-matched children undergoing elective procedures at Oslo University Hospital in the period January 2014 to April 2018 (Table 1). All samples were obtained after informed, written parental consent according to the Declaration of Helsinki and the study approved by the South-Eastern Norway Regional Ethical Committee. Patients were treated according to risk-adapted NOPHO (Nordic Society of Paediatric Haematology and Oncology)–ALL 2008 protocol.29 ALL patients were grouped as non–high risk (non-HR) or high risk (HR) at diagnosis based on the white blood cell count and immunophenotype and were further grouped into standard risk (SR), intermediate risk (IR), or HR based on cytogenetics and minimal residual disease after induction therapy on treatment day 29.30 Blood and BM were consecutively collected from patients at diagnosis (n = 47) and end of treatment (after 130 wk, n = 18). Also, samples were collected during treatment; for BCP-ALL on day 79 following induction and consolidation therapy (n = 42). The samples were immediately processed to isolate mononuclear cells by density gradient centrifugation using Lymphoprep (Axis-Shield), and cells were viably cryopreserved in 20% dimethyl sulfoxide and 80% fetal bovine serum (FBS).

Table 1.

Characteristics of healthy control subjects and patients.

Control subjectsBCP-ALLRisk groups BCP-ALLa
SRIRHR
No.574731 (66%)13 (28%)3 (6%)
Age at diagnosis, y5.0 (0–16)4.0 (1–16)3.0 (1–16)10.0 (2–16)2.0 (2–6)
Male:female ratio21.231.211.60.5
WBC (×109/L)7.9 (0.9–167)7.2 (0.9–92.5)15.3 (1.1–110)19.6 (5.6–167)
CMV (IgG positive)18 (38%)10 (32%)7 (53%)1 (33%)
Cytogenetics BCP-ALL
 SRb15/47141
 IRc1/471
 HRd2/472
 ETV6/RUNX116/47106
 Trisomy 212/472
 Others (including CN)11/47731
Control subjectsBCP-ALLRisk groups BCP-ALLa
SRIRHR
No.574731 (66%)13 (28%)3 (6%)
Age at diagnosis, y5.0 (0–16)4.0 (1–16)3.0 (1–16)10.0 (2–16)2.0 (2–6)
Male:female ratio21.231.211.60.5
WBC (×109/L)7.9 (0.9–167)7.2 (0.9–92.5)15.3 (1.1–110)19.6 (5.6–167)
CMV (IgG positive)18 (38%)10 (32%)7 (53%)1 (33%)
Cytogenetics BCP-ALL
 SRb15/47141
 IRc1/471
 HRd2/472
 ETV6/RUNX116/47106
 Trisomy 212/472
 Others (including CN)11/47731

Abbreviations: ALL = acute lymphoblastic leukemia; BCP-ALL = pre–B cell acute lymphoblastic leukemia; CMV = cytomegalovirus; CN = cytogenetics negative; HR = high risk; Ig = immunoglobulin; IR = intermediate risk; MLL = mixed lineage leukemia ; NOPHO = Nordic Society of Paediatric Haematology and Oncology; SR = standard risk; WBC = white blood cell count.

aDay 29 in NOPHO ALL 2008 treatment protocol.

bCytogenetics: hyperdiploidy.

cCytogenetics: dic(9; 20), t(1; 19), Ic21amp = AML1.

dCytogenetics: MLL (11q23), hypodiploidy.

Table 1.

Characteristics of healthy control subjects and patients.

Control subjectsBCP-ALLRisk groups BCP-ALLa
SRIRHR
No.574731 (66%)13 (28%)3 (6%)
Age at diagnosis, y5.0 (0–16)4.0 (1–16)3.0 (1–16)10.0 (2–16)2.0 (2–6)
Male:female ratio21.231.211.60.5
WBC (×109/L)7.9 (0.9–167)7.2 (0.9–92.5)15.3 (1.1–110)19.6 (5.6–167)
CMV (IgG positive)18 (38%)10 (32%)7 (53%)1 (33%)
Cytogenetics BCP-ALL
 SRb15/47141
 IRc1/471
 HRd2/472
 ETV6/RUNX116/47106
 Trisomy 212/472
 Others (including CN)11/47731
Control subjectsBCP-ALLRisk groups BCP-ALLa
SRIRHR
No.574731 (66%)13 (28%)3 (6%)
Age at diagnosis, y5.0 (0–16)4.0 (1–16)3.0 (1–16)10.0 (2–16)2.0 (2–6)
Male:female ratio21.231.211.60.5
WBC (×109/L)7.9 (0.9–167)7.2 (0.9–92.5)15.3 (1.1–110)19.6 (5.6–167)
CMV (IgG positive)18 (38%)10 (32%)7 (53%)1 (33%)
Cytogenetics BCP-ALL
 SRb15/47141
 IRc1/471
 HRd2/472
 ETV6/RUNX116/47106
 Trisomy 212/472
 Others (including CN)11/47731

Abbreviations: ALL = acute lymphoblastic leukemia; BCP-ALL = pre–B cell acute lymphoblastic leukemia; CMV = cytomegalovirus; CN = cytogenetics negative; HR = high risk; Ig = immunoglobulin; IR = intermediate risk; MLL = mixed lineage leukemia ; NOPHO = Nordic Society of Paediatric Haematology and Oncology; SR = standard risk; WBC = white blood cell count.

aDay 29 in NOPHO ALL 2008 treatment protocol.

bCytogenetics: hyperdiploidy.

cCytogenetics: dic(9; 20), t(1; 19), Ic21amp = AML1.

dCytogenetics: MLL (11q23), hypodiploidy.

2.2 Antibodies and flow cytometry

Antibodies (all anti-human) used were from BD Biosciences, anti-CD8 (SK1-FITC), anti-CD56 (B159-AF647), anti-CD158e1 (DX9-PE), and anti-CD16 (3G8-FITC); from Thermo Fisher Scientific, anti-CD3 (OKT3-AF700), anti-CD14 (M5E2-PerCP-Cy5.5), anti-CD19 (1D3-PerCP-Cy5.5), and anti-NKG2D (A10-biotin), and anti-NKp30 (AF294D12-Pe-Cy7); from R&D Systems, anti-NKG2C (#134591-AF488); from BioLegend, anti-NKp46 (9E2-BV650), anti-DNAM-1 (11.A8-PE), anti-CD4 (RM4-5-PE/Cy7), anti-CD57 (HNK-1-FITC), anti-CXCR3 (G025H7-BV650), anti-CXCR4 (12G5-BV421), and anti-CXCR6 (K041E5-PerCP-Cy5.5); from LifeSpan Biosciences, anti-CD161 (B199.2-biotin); from Beckman Coulter, anti-NKG2A (Z199-PE-Cy7), anti-CD158a, h (EB6-PE), anti-CD158b1/b2, j (GL183-PE), anti-CD158e1/e2 (Z27.3.7-PE), and CD158i (FES172-PE). Streptavidin-QDot605 was purchased from Thermo Fisher Scientific. Cells were labeled with relevant antibodies and analyzed by Fortessa (BD Biosciences). Frozen samples were thawed and rested overnight in complete culture medium (cRPMI) (RPMI supplemented with 10% FBS, 2-mercaptoethanol, sodium pyruvate, and penicillin/streptomycin) prior to analysis. Dead cells were excluded from analysis using the Fixable Viability Dye eFluor780 (eBiosciences). Lymphocytes (singlets) were gated by forward- and side-scatter characteristics, and human NK cells were gated as live CD56+CD3CD14CD19 cells. Data were analyzed using FlowJo version 10.4.1 (TreeStar).

2.3 Degranulation and IFN-γ production

Degranulation by human NK cells were measured by incubating 100 µL peripheral blood mononuclear cells (2 × 106 cells/mL) with 100 µL K562 target cells (2 × 106 cells/mL) in the presence of anti-human CD107a-BV510 (BD Biosciences) for 6 h, with Brefeldin A (Sigma-Aldrich) added at 5 µg/mL for the last 5 h. Cells were fixed with 4% paraformaldehyde for 10 min and permeabilized with 0.5% saponin in phosphate-buffered saline and 2% FBS for 20 min and stained with anti-IFN-γ (BD Biosciences) for 30 min in permeabilization buffer. The human chronic myeloid leukemia cell line K562 (ATCC CCL-243) was maintained in cRPMI.

2.4 Statistical analysis

The data were analyzed using GraphPad Prism (GraphPad Prism 9 Software), R (R Foundation for Statistical Computing, version 4.2.2), and Python (Python Software Foundation, version 3.6.4). Data depicted in graphs are presented as the mean ± SEM. Comparisons within an experimental group of data were performed with 1-way analysis of variance, and with the Mann-Whitney U test to compare data within an experimental group. Unsupervised hierarchical clustering was generated using Euclidean distance. Clustering of markers was performed using the “complete linkage” algorithm using either the R function “hclust”31 or Cluster 3.0 software (ENCODE). Heatmaps were visualized using either the R package NFM.32 Markers with more than 10% missing values were excluded from the analysis, and remaining missing values were imputed using the R package mice.33

3. Results

3.1 Reductions in NK cell degranulation and IFN-γ production at diagnosis and throughout treatment

NK cell effector functions are reportedly reduced in pediatric and adult acute leukemias. We show here that when separately assessing SR and IR patients, NK cell degranulation against sensitive K562 tumor targets was significantly reduced only for the IR group as compared with healthy control subjects and the SR group at diagnosis (Fig. 1A). However, at day 79 degranulation was low for both groups and remained low at the end of treatment. Similarly, IFN-γ production was reduced in NK cells from the IR group at the time of diagnosis (Fig. 1B) but not in the SR patient group. The data thus indicate more functional repression in the IR group. The IR group did not present higher number of blasts at diagnosis than the SR group (Table 1), indicating that the functional suppression may operate through other mechanisms. Only 2 HR group patients were tested, demonstrating low degranulation (average 7% CD107+ NK cells) and low percentage of IFN-γ+ NK cells (average 4.37%) (data not shown).

Reduced NK cell effector functions in intermediate risk BCP-ALL patients. Peripheral blood mononuclear cells from patients or healthy control subjects (healthy donors [HDs]) were co-incubated with K562 target cells in the presence of anti-CD107a monoclonal antibody for 6 h. NK cells were gated as live CD14/CD19−CD3−CD56dim cells. (A) Degranulation was measured as percentage CD107a+ NK cells. (B) Intracellular IFN-γ production was measured as percentage of CD56dim NK cells. The SR group and IR group were analyzed separately. The box boundaries indicates the 25th and 75th percentile, the bars indicate the 90th and 10th percentiles, while the line within the boxes indicates the median. Data are presented as percentage ± SEM. Statistical significance was calculated using the nonparametrical Mann-Whitney test. *P < 0.05, **P < 0.01.
Fig. 1.

Reduced NK cell effector functions in intermediate risk BCP-ALL patients. Peripheral blood mononuclear cells from patients or healthy control subjects (healthy donors [HDs]) were co-incubated with K562 target cells in the presence of anti-CD107a monoclonal antibody for 6 h. NK cells were gated as live CD14/CD19CD3CD56dim cells. (A) Degranulation was measured as percentage CD107a+ NK cells. (B) Intracellular IFN-γ production was measured as percentage of CD56dim NK cells. The SR group and IR group were analyzed separately. The box boundaries indicates the 25th and 75th percentile, the bars indicate the 90th and 10th percentiles, while the line within the boxes indicates the median. Data are presented as percentage ± SEM. Statistical significance was calculated using the nonparametrical Mann-Whitney test. *P < 0.05, **P < 0.01.

3.2 Reductions in NKG2D and DNAM-1 expression levels at diagnosis were further exacerbated throughout treatment

Next, expression of activating and inhibitory NK cell receptors was assessed in matched blood and BM samples at the time of diagnosis, during treatment, and at the end of treatment compared with blood and BM from age-matched healthy pediatric control subjects. The frequency of DNAM-1+ NK cells was high on both BM and blood NK cells at diagnosis and comparable to control subjects (Fig. 2A), but the expression levels were greatly reduced on CD56bright and CD56dim NK cells in both SR and IR groups compared with control subjects (Fig. 2B). While normalizing in blood at end of treatment, the frequencies of DNAM-1 remained low in BM. As only 3 HR group patients were included in the study, the phenotypic data were not shown, but tended to accentuate the phenotype of the IR group for all tested markers (data not shown).

Reduced expression levels of DNAM-1 and NKG2D and increased levels of NKp30 at diagnosis. Box plots showing frequencies or expression levels (relative fluorescence intensity [RFI], median fluorescence intensity of marker/median fluorescence intensity of negative control) of DNAM-1 (A, B), NKG2D (C, D), and NKp30 (E, F) by BM or blood CD56bright (top row) or CD56dim (bottom row) NK cells in BCP-ALL patients compared with healthy pediatric control subjects (healthy donors [HDs]). The SR group and the IR group was analysed separately . The box boundaries indicates the 25th and 75th percentile, the bars indicate the 90th and 10th percentiles, and the line within the boxes indicates the median. Statistical significance was calculated by parametrical 1-way analysis of variance. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 2.

Reduced expression levels of DNAM-1 and NKG2D and increased levels of NKp30 at diagnosis. Box plots showing frequencies or expression levels (relative fluorescence intensity [RFI], median fluorescence intensity of marker/median fluorescence intensity of negative control) of DNAM-1 (A, B), NKG2D (C, D), and NKp30 (E, F) by BM or blood CD56bright (top row) or CD56dim (bottom row) NK cells in BCP-ALL patients compared with healthy pediatric control subjects (healthy donors [HDs]). The SR group and the IR group was analysed separately . The box boundaries indicates the 25th and 75th percentile, the bars indicate the 90th and 10th percentiles, and the line within the boxes indicates the median. Statistical significance was calculated by parametrical 1-way analysis of variance. *P < 0.05, **P < 0.01, ***P < 0.001.

We observed no major changes in the frequencies of NKG2D+ NK cells at the time of diagnosis in neither BM nor blood, except a tendency for lower frequencies at end of treatment (Fig. 2C). However, like DNAM-1, NKG2D expression levels were reduced at diagnosis in CD56bright and CD56dim NK cells (Fig. 2D) and remained low in the IR group at the end of treatment on both BM and blood NK cells.

We observed highly variable frequencies of NKp30+ NK cells, but NKp30 frequencies were increased as compared with control subjects for CD56bright BM and blood NK cells at the time of diagnosis (Fig. 2E). Also, NKp30 expression levels were increased at the time of diagnosis in both CD56bright and CD56dim blood NK cells (Fig. 2F). The expression levels of NKp30 tended to stay elevated within the IR group. The expression levels and frequencies of CD16 and NKp46 were not significantly altered in NK cells in any of the patient groups at the time of diagnosis, but fell sharply by the end of treatment (data not shown).

3.3 High NKG2A expression levels at diagnosis remain high throughout treatment

The frequencies of NKG2A+ NK cells were largely comparable to healthy control subjects at the time of diagnosis, with a reduction in blood for the IR group at diagnosis (Fig. 3A). At end of treatment, we observed higher frequencies of NKG2A+ CD56dim BM and blood NK cells within the IR group. At diagnosis, the NKG2A expression levels were increased at the time of diagnosis in blood CD56bright and CD56dim NK cells, as well as in BM CD56dim NK cells for the IR group (Fig. 3B). The NKG2A expression levels generally remained elevated throughout treatment.

Increased expression levels of NKG2A in BCP-ALL patients at diagnosis and throughout treatment. Box plots showing frequencies or expression levels (relative fluorescence intensity [RFI], median fluorescence intensity of marker/median fluorescence intensity of negative control) of NKG2A (A, B) or CD161 (C, D) by BM or blood CD56bright (top row) or CD56dim (bottom row) NK cells. The SR group and the IR group was analysed separately. The box boundaries indicates the 25th and 75th percentile, the bars indicate the 90th and 10th percentiles, while the line within the boxes indicates the median. Statistical significance was calculated by parametrical 1-way analysis of variance. *P < 0.05, **P < 0.01, ***P < 0.001. HD = healthy donor.
Fig. 3.

Increased expression levels of NKG2A in BCP-ALL patients at diagnosis and throughout treatment. Box plots showing frequencies or expression levels (relative fluorescence intensity [RFI], median fluorescence intensity of marker/median fluorescence intensity of negative control) of NKG2A (A, B) or CD161 (C, D) by BM or blood CD56bright (top row) or CD56dim (bottom row) NK cells. The SR group and the IR group was analysed separately. The box boundaries indicates the 25th and 75th percentile, the bars indicate the 90th and 10th percentiles, while the line within the boxes indicates the median. Statistical significance was calculated by parametrical 1-way analysis of variance. *P < 0.05, **P < 0.01, ***P < 0.001. HD = healthy donor.

CD161 (NKR-P1A) was not significantly altered at diagnosis, with the exception of higher frequencies by CD56bright BM and blood NK cells within the IR group (Fig. 3C). Sharp reductions of CD161 frequencies were observed for both BM and blood NK cells at end of treatment. No significant changes in the expression levels of CD161 were observed (Fig. 3D).

3.4 High expression levels of NKG2A discriminates NK cells in patients from healthy control subjects

Substantial interindividual variations were observed in receptor frequencies and expression levels. Nonetheless, the data indicated an overall skewing toward decreased expression levels of activating receptors and increased expression levels of NKG2A. We performed an unsupervised hierarchical cluster analysis of 11 surface markers and of intracellular perforin to identify potential aberrant phenotypic signatures at the time of diagnosis. NKp30 and CD16 was excluded from this analysis due to more than 10% missing values. Two main clusters were observed, with healthy control subjects (in green) generally clustering away from patients (in blue) for CD56dim blood NK cells (Fig. 4A). The partitioning was driven by a skewing of individuals with very high expression levels of NKG2A compared with individuals with high frequencies of NKp46, NKG2D, DNAM-1, CD161, and perforin. Moreover, there was an enrichment of IR group patients within the NKG2Ahigh cluster, as well as 3 of 4 patients that had relapsed within the time frame of our study. No clustering with regard to cytogenetics was observed (data not shown). For this analysis, the HR patients were included, and it was clear that these 3 patients had a quite divergent profile with respect to NKG2A and activating receptor expression levels. In the BM, a similar segregation of patients was observed, but the number of healthy control BM samples was too low for meaningful comparison (data not shown).

High expression levels of NKG2A discriminates NK cells in patients from healthy control subjects (healthy donors [HDs]). (A) Heatmaps showing unsupervised hierarchical clustering of phenotypic markers on CD56dim blood NK cells. Columns represents individual healthy control subjects or patients. Rows represent normalized levels of expression for an indicated phenotypic parameter. Risk categories are indicated for each individual patient. (B) Frequencies of NKG2A/CD57 subsets within healthy pediatric control subjects or BCP-ALL patients at diagnosis. Statistical significance was calculated using the nonparametrical Mann-Whitney test. NA= not applicable.
Fig. 4.

High expression levels of NKG2A discriminates NK cells in patients from healthy control subjects (healthy donors [HDs]). (A) Heatmaps showing unsupervised hierarchical clustering of phenotypic markers on CD56dim blood NK cells. Columns represents individual healthy control subjects or patients. Rows represent normalized levels of expression for an indicated phenotypic parameter. Risk categories are indicated for each individual patient. (B) Frequencies of NKG2A/CD57 subsets within healthy pediatric control subjects or BCP-ALL patients at diagnosis. Statistical significance was calculated using the nonparametrical Mann-Whitney test. NA= not applicable.

To further assess NKG2A expression, we next assessed the maturation stage of NK cells in blood at the time of diagnosis. The least mature CD57NKG2A+ CD56dim NK cells was the most dominant subset observed in both healthy children and patients (Fig. 4B). For patients, we observed a heterogeneity of maturational stages in BM and blood NK cells during treatment, but by end of treatment NK cells had a predominant naïve CD57NKG2A+/− phenotype (data not shown).

3.5 Higher fractions of hypofunctional CXCR6+DNAM-1negNKG2A+ NK cells in leukemia patients

Finally, we tested whether the leukemia would affect tissue-resident NK cells of the BM. The BM contains a distinct population of resident NK cells that uniquely expresses CD69 and the chemokine receptor CXCR6, with both markers largely absent on circulating NK cells.20 We show here that CXCR6+ BM NK cells are found in similar frequencies in healthy control subjects and BCP-ALL patients (Fig. 5A and B). The CXCR6+ NK cells are hypofunctional compared with CD56dim NK cells, as measured as degranulation against K562 targets (Fig. 5C). A proportion of CXCR6+ NK cells lack DNAM-1 expression, and the lack of DNAM-1 is further accentuated in patients (Fig. 5D and E). The reduced frequency of DNAM-1 by CXCR6+ NK cells in the patients may contribute to further loss of cytotoxic capacity by this subset, although this was not significant with the small number of samples tested (Fig. 5F). Further, we show that the DNAM-1 negative fraction of CXCR6+ NK cells express proportionally more NKG2A in patients compared with healthy control subjects (Fig. 5G and H).

Higher frequencies of CXCR6+DNAM-1negNKG2A+ BM NK cells in BCP-ALL patients. (A) Representative flow cytometry plots showing of CXCR6 + CD69+ BM NK cells in healthy donors (HDs) or BCP-ALL patients. (B) Frequencies of CXCR6+CD69+ BM NK cells in HDs (n = 10) or BCP-ALL patients at diagnosis according to their risk status: SR (n = 25), IR (n = 13). Data are presented as percentage ± SEM. (C) Degranulation of CD56dim or CXCR6+ BM NK cells after co-incubation of BM mononuclear cells with K562 target cells for 4 h. (D) DNAM-1 expression by BM NK cell subsets in HDs or BCP-ALL patients. (E) Frequencies of DNAM-1neg CXCR6+ NK cells in HDs (n = 10) vs BCP-ALL patients (n = 38). (F) Degranulation of DNAM-1posCXCR6+ vs DNAM-1negCXCR6+ BM NK cells in HDs (n = 4) vs BCP-ALL patients (n = 7). (G, H) Frequencies of DNAM-1/NKG2A subsets in HDs (n = 3) vs BCP-ALL patients (n = 6). Data presented as percentage ± SEM, statistical significance was calculated using the nonparametrical Mann-Whitney test.
Fig. 5.

Higher frequencies of CXCR6+DNAM-1negNKG2A+ BM NK cells in BCP-ALL patients. (A) Representative flow cytometry plots showing of CXCR6 + CD69+ BM NK cells in healthy donors (HDs) or BCP-ALL patients. (B) Frequencies of CXCR6+CD69+ BM NK cells in HDs (n = 10) or BCP-ALL patients at diagnosis according to their risk status: SR (n = 25), IR (n = 13). Data are presented as percentage ± SEM. (C) Degranulation of CD56dim or CXCR6+ BM NK cells after co-incubation of BM mononuclear cells with K562 target cells for 4 h. (D) DNAM-1 expression by BM NK cell subsets in HDs or BCP-ALL patients. (E) Frequencies of DNAM-1neg CXCR6+ NK cells in HDs (n = 10) vs BCP-ALL patients (n = 38). (F) Degranulation of DNAM-1posCXCR6+ vs DNAM-1negCXCR6+ BM NK cells in HDs (n = 4) vs BCP-ALL patients (n = 7). (G, H) Frequencies of DNAM-1/NKG2A subsets in HDs (n = 3) vs BCP-ALL patients (n = 6). Data presented as percentage ± SEM, statistical significance was calculated using the nonparametrical Mann-Whitney test.

4. Discussion

In this study, we performed a comparative profiling of BM and blood NK cells from pediatric BCP-ALL patients compared with heathy pediatric control subjects. Our data demonstrate large variations in receptor expression between individuals, in both control subjects and patients. The variations were age independent (data not shown), in line with a previous study.34 By integrating hierarchical clustering of the phenotypic markers tested in our study, we show that patients and healthy control subjects segregate mainly due to disparately high expression of NKG2A vs lower expression of NKp46, DNAM-1, NKG2D, and CD161 in patients.

In line with previous reports,15,35 we observed generally suppressed features of NK cells at diagnosis with skewing toward less expression of activating NK cell receptors and low effector functions that remained throughout treatment. In particular, surface expression of the activating receptors DNAM-1 and NKG2D were reduced on the NK cells in patients. Reduced surface expression of receptors may occur via ligand-induced receptor downregulation. We did not systematically test ligand expression by the leukemic blasts, but BCP-ALL have previously been shown to express the DNAM-1 ligand CD112 and NKG2D ligands,9,36 which could explain the findings. In contrast to previous studies,8,15 we found no changes in NKp46 expression at diagnosis, but NKp46 expression levels were very variable between individuals.

In contrast to DNAM-1 and NKG2D, we observed a general upregulation of NKp30 expression levels at diagnosis. Low NKp30 levels were in a previous study correlated to increased relapse risk for adult AML.37 For BCP-ALL, a previous study found no changes in NKp30 expression by NK cells.15 High expression levels of NKp30 may seem counterintuitive, but recent research indicates that 3 isoforms exist for NKp30. While NKp30a/b induces NK cell activation, the NKp30 isoform mediates NK cell inhibitory effects through secretion of interleukin-10.38 We speculate that the patients potentially could overexpress the NKp30c isoform at diagnosis, but this remains to be tested.

The majority of patients had NK cells with high levels of NKG2A at diagnosis compared with healthy control subjects. This finding is in line with a previous study reporting increased expression levels of NKG2A by NK cells in pediatric ALL patients at diagnosis.15 Increased levels of NKG2A may inhibit NK cells through interactions with HLA-E. Several cancers overexpress HLA-E, including hematological cancers.39,40 HLA-E is expressed at low levels in pediatric ALL blasts, and it has been suggested that the downmodulation is the result of active immunosurveillance by NKG2A+ NK cells.41 Blocking NKG2A with the humanized anti-NKG2A antibody monalizumab has shown promise as a means to prevent NKG2A inhibitory signaling, has been shown to improve NK cell–mediated killing of HLA-E+ chronic lymphocytic leukemia in vitro, and has improved response rates of colorectal cancer and head and neck cancer.40,42,43 Other approaches to target NKG2A include blockers designed to prevent surface expression of NKG2A, which yields NK cells with heightened activity.44 With regard to BCP-ALL, NKG2A blockade may still unleash NK cell cytotoxicity despite the low HLA-E levels, as there may still be a functional interaction.

We observed low NK cell functional activity in the IR group at diagnosis. It has been suggested that low HLA-E expression levels could lead to failure of NK cell education, driving NK cells into a hyporesponsive state,41 which potentially could explain the observed low functionality of NK cells. Alternatively, it was previously proposed that the low NK cell activity present at diagnosis results from the suppressive activity of transforming growth factor β released from the blasts.15 In this respect, we observed a weak correlation of lower NK cell activity with higher blast counts (data not shown). The latter could represent an explanation for the normal functional activity in the SR group at diagnosis, and the fact that we did not observe major differences in NK cell receptor expression between the SR and IR groups.

Expression levels of the inhibitory receptor CD161 were decreased throughout treatment. While we found no alterations in CD161 in the patient group as a whole, we did find low levels of CD161 in NK cells that concomitantly expressed high levels of NKG2A. The functional role of CD161 on NK cells is still unclear, and it is even suggested to have an activating function in T cells.45 It is unclear whether human CD161 is implicated in targeting of leukemic blasts via its ligand LLT1, but the inhibitory NKR-P1B receptor and its ligand Clr-b have been implicated in tumor immune surveillance in mice.46

The chemotherapy regimens are likely a contributor to observed changes in receptor expression throughout treatment as well as to the reduced NK cell activity observed at day 79, when the presence of blasts is minimal. We observed drops in frequencies of NKG2D+, DNAM-1+, and NKp46+ NK cells from day 79 onward, which was not evident at diagnosis. Several clinical studies have documented decreased NK cell activity or changes in activating receptors in conjunction with different chemotherapies. Relevant for our study, corticosteroid treatment of pediatric patients was shown to reduce surface expression of NKG2D, NKp30, and NKp46 and lower the activity of NK cells.47,48 It is thus difficult to conclude whether the phenotypic changes observed at day 79 are due to intrinsic defects in the NK cells or to an effect of the chemotherapeutic regimen. By the end of treatment, we observed a higher abundance of NKG2A+CD57KIR immature NK cells (data not shown), indicating that newly developed NK cells may dominate the circulating pool of NK cells at this point.

We found changes in the proportions of mature vs naïve NK cells in patients compared with control subjects at diagnosis, with more naïve NKG2A+CD57 NK cells in patients, suggesting that the leukemia affects NK cell differentiation, directly or indirectly, and may impair development of cytolytically competent NK cells. Our data align with a recent study on a cohort of Mexican BCP-ALL patients in which similar findings were made.35 Cytomegalovirus infection leads to imprinting of the NK cell receptor repertoire,49 linked to the generation of adaptive CD56dimCD57+NKG2C+ NK cells, and there is evidence for a more efficient antileukemic effect with these cells.50 However, a patient with high fractions of putative adaptive NK cells (data not shown) at end of treatment in our cohort relapsed shortly after.

CXCR6+ tissue-resident NK cells are reportedly hypofunctional and express lower levels of granzyme B and perforin.20 We also found that CXCR6+ NK cells have lower capacity for degranulation compared with CD56dim NK cells. Moreover, we found that a large fraction of CXCR6+ NK cells in patients lack expression of the activating receptor DNAM-1. DNAM-1 is generally expressed by all circulating NK cells, while expressed in a bimodal fashion on BM CXCR6+ NK cells.20 The DNAM-1–negative fraction was larger in patients than in healthy control subjects, irrespective of risk groups, indicating that this phenotype is a general feature of NK cells from BCP-ALL patients. Moreover, the DNAM-1neg fraction in patients had lower degranulation capacity compared with the DNAM-1pos fraction. The lower expression of DNAM-1 by CXCR6+ NK cells in patients may be related to ligand-induced receptor downregulation. The DNAM-1 ligands CD155 and CD112 are variably expressed in hematological malignancies and are directly linked to receptor downregulation.3,12 However, this does not fully explain the existence of DNAM-1neg cells, as around 40% of CXCR6+ NK cells of healthy individuals also lack DNAM-1. Possibly, DNAM-1negCXCR6+ NK cells could represent a separate subset with distinct functions (i.e. expanded in patients irrespective of DNAM-1 ligands). The BM of ALL patients are also reported to harbor a suppressive CD56bright NK cell subset producing transforming growth factor β and interleukin-10 that could contribute to pathology.51 These cells expressed lower levels of activating receptors reminiscent to the CD56bright NK cells profiled in our study.

Conclusions

Our study highlights that multiparameter analyses of NK cells is necessary to reveal the full phenotypic dimensional space of NK cells in patients due to large interindividual variations in phenotypes. We identified a profile dominated by high expression levels of NKG2A that marked a patient group with a more suppressed phenotype and suggest that NKG2A could be targeted in therapies of pediatric acute leukemia.

Acknowledgments

The authors thank the children, donors, and parents involved in this study. This work was supported by the South-Eastern Regional Health Authority under grant 2013068, Fondsstiftelsen at Oslo University Hospital, and UNIFOR-FRIMED.

Author contributions

A.U., A.G.B., and M.I. conceptualized and designed the study; A.U. and M.I. wrote the manuscript; A.U. and M.I. conducted experiments; A.U., M.I., V.G., and A.G.B. analyzed and interpreted the data; all authors read, edited and approved the final manuscript.

References

1

Toft
N
,
Birgens
H
,
Abrahamsson
J
,
Griskevicius
L
,
Hallbook
H
,
Heyman
M
,
Klausen
TW
,
Jonsson
OG
,
Palk
K
,
Pruunsild
K
, et al.
Results of NOPHO ALL2008 treatment for patients aged 1-45 years with acute lymphoblastic leukemia
.
Leukemia
.
2018
:
32
(
3
):
606
615
. https://doi.org/10.1038/leu.2017.265

2

Boieri
M
,
Ulvmoen
A
,
Sudworth
A
,
Lendrem
C
,
Collin
M
,
Dickinson
AM
,
Kveberg
L
,
Inngjerdingen
M
.
IL-12, IL-15, and IL-18 pre-activated NK cells target resistant T cell acute lymphoblastic leukemia and delay leukemia development in vivo
.
Oncoimmunology
.
2017
:
6
(
3
):
e1274478
. https://doi.org/10.1080/2162402X.2016.1274478

3

Carlsten
M
,
Baumann
BC
,
Simonsson
M
,
Jädersten
M
,
Forsblom
AM
,
Hammarstedt
C
,
Bryceson
YT
,
Ljunggren
HG
,
Hellström-Lindberg
E
,
Malmberg
KJ
.
Reduced DNAM-1 expression on bone marrow NK cells associated with impaired killing of CD34+blasts in myelodysplastic syndrome
.
Leukemia
.
2010
:
24
(
9
):
1607
1616
. https://doi.org/10.1038/leu.2010.149

4

Sørskaar
D
,
Lie
SO
,
Førre
O
.
Natural killer cell activity of peripheral blood and bone marrow mononuclear cells from patients with childhood acute lymphoblastic leukemia
.
Acta Paediatr Scand
.
1985
:
74
(
3
):
433
437
. https://doi.org/10.1111/j.1651-2227.1985.tb10998.x

5

Stringaris
K
,
Sekine
T
,
Khoder
A
,
Alsuliman
A
,
Razzaghi
B
,
Sargeant
R
,
Pavlu
J
,
Brisley
G
,
de Lavallade
H
,
Sarvaria
A
, et al.
Leukemia-induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia
.
Haematologica
.
2014
:
99
(
5
):
836
847
. https://doi.org/10.3324/haematol.2013.087536

6

Tajima
F
,
Kawatani
T
,
Endo
A
,
Kawasaki
H
.
Natural killer cell activity and cytokine production as prognostic factors in adult acute leukemia
.
Leukemia
.
1996
:
10
:
478
482
.

7

Lowdell
MW
,
Craston
R
,
Samuel
D
,
Wood
ME
,
O'Neill
E
,
Saha
V
,
Prentice
HG
.
Evidence that continued remission in patients treated for acute leukaemia is dependent upon autologous natural killer cells
.
Br J Haematol
.
2002
:
117
(
4
):
821
827
. https://doi.org/10.1046/j.1365-2141.2002.03495.x

8

Costello
RT
,
Sivori
S
,
Marcenaro
E
,
Lafage-Pochitaloff
M
,
Mozziconacci
MJ
,
Reviron
D
,
Gastaut
JA
,
Pende
D
,
Olive
D
,
Moretta
A
.
Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia
.
Blood
.
2002
:
99
(
10
):
3661
3667
. https://doi.org/10.1182/blood.V99.10.3661

9

Pende
D
,
Spaggiari
GM
,
Marcenaro
S
,
Martini
S
,
Rivera
P
,
Capobianco
A
,
Falco
M
,
Lanino
E
,
Pierri
I
,
Zambello
R
, et al.
Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the poliovirus receptor (CD155) and nectin-2 (CD112)
.
Blood
.
2005
:
105
(
5
):
2066
2073
. https://doi.org/10.1182/blood-2004-09-3548

10

Pende
D
,
Cantoni
C
,
Rivera
P
,
Vitale
M
,
Castriconi
R
,
Marcenaro
S
,
Nanni
M
,
Biassoni
R
,
Bottino
C
,
Moretta
A
, et al.
Role of NKG2D in tumor cell lysis mediated by human NK cells: cooperation with natural cytotoxicity receptors and capability of recognizing tumors of nonepithelial origin
.
Eur J Immunol
.
2001
:
31
(
4
):
1076
1086
. https://doi.org/10.1002/1521-4141(200104)31:4<1076::AID-IMMU1076>3.0.CO;2-Y

11

Fauriat
C
,
Just-Landi
S
,
Mallet
F
,
Arnoulet
C
,
Sainty
D
,
Olive
D
,
Costello
RT
.
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
330
. https://doi.org/10.1182/blood-2005-08-027979

12

Sanchez-Correa
B
,
Morgado
S
,
Gayoso
I
,
Bergua
JM
,
Casado
JG
,
Arcos
MJ
,
Bengochea
ML
,
Duran
E
,
Solana
R
,
Tarazona
R
.
Human NK cells in acute myeloid leukaemia patients: analysis of NK cell-activating receptors and their ligands
.
Cancer Immunol Immunother
.
2011
:
60
(
8
):
1195
1205
. https://doi.org/10.1007/s00262-011-1050-2

13

Hilpert
J
,
Grosse-Hovest
L
,
Grünebach
F
,
Buechele
C
,
Nuebling
T
,
Raum
T
,
Steinle
A
,
Salih
HR
.
Comprehensive analysis of NKG2D ligand expression and release in leukemia: implications for NKG2D-mediated NK cell responses
.
J Immunol
.
2012
:
189
(
3
):
1360
1371
. https://doi.org/10.4049/jimmunol.1200796

14

Khaznadar
Z
,
Boissel
N
,
Agaugue
S
,
Henry
G
,
Cheok
M
,
Vignon
M
,
Geromin
D
,
Cayuela
JM
,
Castaigne
S
,
Pautas
C
, et al.
Defective NK cells in acute myeloid leukemia patients at diagnosis are associated with blast transcriptional signatures of immune evasion
.
J Immunol
.
2015
:
195
(
6
):
2580
2590
. https://doi.org/10.4049/jimmunol.1500262

15

Rouce
RH
,
Shaim
H
,
Sekine
T
,
Weber
G
,
Ballard
B
,
Ku
S
,
Barese
C
,
Murali
V
,
Wu
MF
,
Liu
H
, et al.
The TGF-beta/SMAD pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia
.
Leukemia
.
2016
:
30
(
4
):
800
811
. https://doi.org/10.1038/leu.2015.327

16

Björkström
NK
,
Riese
P
,
Heuts
F
,
Andersson
S
,
Fauriat
C
,
Ivarsson
MA
,
Björklund
AT
,
Flodström-Tullberg
M
,
Michaëlsson
J
,
Rottenberg
ME
, et al.
Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education
.
Blood
.
2010
:
116
(
19
):
3853
3864
. https://doi.org/10.1182/blood-2010-04-281675

17

Lopez-Verges
S
,
Milush
JM
,
Pandey
S
,
York
VA
,
Arakawa-Hoyt
J
,
Pircher
H
,
Norris
PJ
,
Nixon
DF
,
Lanier
LL
.
CD57 Defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset
.
Blood
.
2010
:
116
(
19
):
3865
3874
. https://doi.org/10.1182/blood-2010-04-282301

18

Chretien
AS
,
Fauriat
C
,
Orlanducci
F
,
Galseran
C
,
Rey
J
,
Bouvier Borg
G
,
Gautherot
E
,
Granjeaud
S
,
Hamel-Broza
JF
,
Demerle
C
, et al.
Natural killer defective maturation is associated with adverse clinical outcome in patients with acute myeloid leukemia
.
Front Immunol
.
2017
:
8
:
573
. https://doi.org/10.3389/fimmu.2017.00573

19

Björklund
AT
,
Clancy
T
,
Goodridge
JP
,
Beziat
V
,
Schaffer
M
,
Hovig
E
,
Ljunggren
HG
,
Ljungman
PT
,
Malmberg
KJ
.
Naive donor NK cell repertoires associated with less leukemia relapse after allogeneic hematopoietic stem cell transplantation
.
J Immunol
.
2016
:
196
(
3
):
1400
1411
. https://doi.org/10.4049/jimmunol.1501434

20

Lugthart
G
,
Melsen
JE
,
Vervat
C
,
van Ostaijen-Ten Dam
MM
,
Corver
WE
,
Roelen
DL
,
van Bergen
J
,
van Tol
MJ
,
Lankester
AC
,
Schilham
MW
.
Human lymphoid tissues harbor a distinct CD69+CXCR6+ NK cell population
.
J Immunol
.
2016
:
197
(
1
):
78
84
. https://doi.org/10.4049/jimmunol.1502603

21

Shiow
LR
,
Rosen
DB
,
Brdickova
N
,
Xu
Y
,
An
J
,
Lanier
LL
,
Cyster
JG
,
Matloubian
M
.
CD69 Acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs
.
Nature
.
2006
:
440
(
7083
):
540
544
. https://doi.org/10.1038/nature04606

22

Rebuli
ME
,
Pawlak
EA
,
Walsh
D
,
Martin
EM
,
Jaspers
I
.
Distinguishing human peripheral blood NK cells from CD56dimCD16dimCD69+CD103+ resident nasal mucosal lavage fluid cells
.
Sci Rep
.
2018
:
8
(
1
):
3394
. https://doi.org/10.1038/s41598-018-21443-5

23

Marquardt
N
,
Kekäläinen
E
,
Chen
P
,
Kvedaraite
E
,
Wilson
JN
,
Ivarsson
MA
,
Mjösberg
J
,
Berglin
L
,
Säfholm
J
,
Manson
ML
, et al.
Human lung natural killer cells are predominantly comprised of highly differentiated hypofunctional CD69-CD56dim cells
.
J Allergy Clin Immunol
.
2017
:
1139
:
321
1330.e4
. https://doi.org/10.1016/j.jaci.2016.07.043

24

Montaldo
E
,
Vacca
P
,
Chiossone
L
,
Croxatto
D
,
Loiacono
F
,
Martini
S
,
Ferrero
S
,
Walzer
T
,
Moretta
L
,
Mingari
MC
.
Unique eomes+ NK cell subsets are present in uterus and decidua during early pregnancy
.
Front Immunol
.
2015
:
6
:
646
. https://doi.org/10.3389/fimmu.2015.00646

25

Marquardt
N
,
Béziat
V
,
Nyström
S
,
Hengst
J
,
Ivarsson
MA
,
Kekäläinen
E
,
Johansson
H
,
Mjösberg
J
,
Westgren
M
,
Lankisch
TO
, et al.
Cutting edge: identification and characterization of human intrahepatic CD49a+ NK cells
.
J Immunol
.
2015
:
194
(
6
):
2467
2471
. https://doi.org/10.4049/jimmunol.1402756

26

Cuff
AO
,
Robertson
FP
,
Stegmann
KA
,
Pallett
LJ
,
Maini
MK
,
Davidson
BR
,
Male
V
.
Eomeshi NK cells in human liver are long-lived and do not recirculate but can be replenished from the circulation
.
J Immunol
.
2016
:
197
(
11
):
4283
4291
. https://doi.org/10.4049/jimmunol.1601424

27

Stegmann
KA
,
Robertson
F
,
Hansi
N
,
Gill
U
,
Pallant
C
,
Christophides
T
,
Pallett
LJ
,
Peppa
D
,
Dunn
C
,
Fusai
G
, et al.
CXCR6 Marks a novel subset of T-betloEomeshi natural killer cells residing in human liver
.
Sci Rep
.
2016
:
6
(
1
):
26157
. https://doi.org/10.1038/srep26157

28

Collins
A
,
Rothman
N
,
Liu
K
,
Reiner
SL
.
Eomesodermin and T-bet mark developmentally distinct human natural killer cells
.
JCI insight
.
2017
:
2
(
5
):
e90063
. https://doi.org/10.1172/jci.insight.90063

29

Raja
RA
,
Schmiegelow
K
,
Albertsen
BK
,
Prunsild
K
,
Zeller
B
,
Vaitkeviciene
G
,
Abrahamsson
J
,
Heyman
M
,
Taskinen
M
,
Harila-Saari
A
, et al.
Asparaginase-associated pancreatitis in children with acute lymphoblastic leukaemia in the NOPHO ALL2008 protocol
.
Br J Haematol
.
2014
:
165
(
1
):
126
133
. https://doi.org/10.1111/bjh.12733

30

Toft
N
,
Birgens
H
,
Abrahamsson
J
,
Bernell
P
,
Griskevicius
L
,
Hallbook
H
,
Heyman
M
,
Holm
MS
,
Hulegardh
E
,
Klausen
TW
, et al.
Risk group assignment differs for children and adults 1-45 yr with acute lymphoblastic leukemia treated by the NOPHO ALL-2008 protocol
.
Eur J Haematol
.
2013
:
90
(
5
):
404
412
. https://doi.org/10.1111/ejh.12097

31

Chang
JT
,
Gatza
ML
,
Lucas
JE
,
Barry
WT
,
Vaughn
P
,
Nevins
JR
.
SIGNATURE: a workbench for gene expression signature analysis
.
BMC Bioinformatics
.
2011
:
12
:
443
. https://doi.org/10.1186/1471-2105-12-443

32

Gaujoux
R
,
Seoighe
C
.
A flexible R package for nonnegative matrix factorization
.
BMC Bioinformatics
.
2010
:
11
:
367
. https://doi.org/10.1186/1471-2105-11-367

33

van Buuren
S
,
Groothuis-Oudshoorn
K
.
Mice: multivariate imputation by chained equations in R
.
J Stat Softw
.
2011
:
45(3)
:
1–67
.

34

Mahapatra
S
,
Mace
EM
,
Minard
CG
,
Forbes
LR
,
Vargas-Hernandez
A
,
Duryea
TK
,
Makedonas
G
,
Banerjee
PP
,
Shearer
WT
,
Orange
JS
.
High-resolution phenotyping identifies NK cell subsets that distinguish healthy children from adults
.
PLoS One
.
2017
:
12
(
8
):
e0181134
. https://doi.org/10.1371/journal.pone.0181134

35

Valenzuela-Vázquez
L
,
Nuñez-Enriquez
JC
,
Sánchez-Herrera
J
,
Medina-Sanson
A
,
Pérez-Saldivar
ML
,
Jiménez-Hernández
E
,
Martiín-Trejo
JA
,
Campo-Martínez
D
,
de Los Ángeles
M
,
Flores-Lujano
J
.
NK Cells with decreased expression of multiple activating receptors is a dominant phenotype in pediatric patients with acute lymphoblastic leukemia
.
Front Oncol
.
2022
:
12
:
1023510
. https://doi.org/10.3389/fonc.2022.1023510

36

Torelli
GF
,
Peragine
N
,
Raponi
S
,
Pagliara
D
,
De Propris
MS
,
Vitale
A
,
Bertaina
A
,
Barberi
W
,
Moretta
L
,
Basso
G
, et al.
Recognition of adult and pediatric acute lymphoblastic leukemia blasts by natural killer cells
.
Haematologica
.
2014
:
99
(
7
):
1248
1254
. https://doi.org/10.3324/haematol.2013.101931

37

Chretien
AS
,
Fauriat
C
,
Orlanducci
F
,
Rey
J
,
Borg
GB
,
Gautherot
E
,
Granjeaud
S
,
Demerle
C
,
Hamel
JF
,
Cerwenka
A
, et al.
NKp30 expression is a prognostic immune biomarker for stratification of patients with intermediate-risk acute myeloid leukemia
.
Oncotarget
.
2017
:
8
(
30
):
49548
49563
. https://doi.org/10.18632/oncotarget.17747

38

Delahaye
NF
,
Rusakiewicz
S
,
Martins
I
,
Ménard
C
,
Roux
S
,
Lyonnet
L
,
Paul
P
,
Sarabi
M
,
Chaput
N
,
Semeraro
M
, et al.
Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors
.
Nat Med
.
2011
:
17
(
6
):
700
707
. https://doi.org/10.1038/nm.2366

39

Márin
R
,
Ruiz-Cabello
F
,
Pedrinaci
S
,
Méndez
R
,
Jiménez
P
,
Geraghty
DE
,
Garrido
F
.
Analysis of HLA-E expression in human tumors
.
Immunogenetics
.
2003
:
54
(
11
):
767
775
. https://doi.org/10.1007/s00251-002-0526-9

40

Ruggeri
L
,
Urbani
E
,
André
P
,
Mancusi
A
,
Tosti
A
,
Topini
F
,
Bléry
M
,
Animobono
L
,
Romagné
F
,
Wagtmann
N
, et al.
Effects of anti-NKG2A antibody administration on leukemia and normal hematopoietic cells
.
Haematologica
.
2016
:
101
(
5
):
626
633
. https://doi.org/10.3324/haematol.2015.135301

41

Reusing
SB
,
Manser
AR
,
Enczmann
J
,
Mulder
A
,
Claas
FH
,
Carrington
M
,
Fischer
JC
,
Borkhardt
A
,
Babor
F
,
Uhrberg
M
.
Selective downregulation of HLA-C and HLA-E in childhood acute lymphoblastic leukaemia
.
Br J Haematol
.
2016
:
174
(
3
):
477
480
. https://doi.org/10.1111/bjh.13777

42

McWilliams
EM
,
Mele
JM
,
Cheney
C
,
Timmerman
EA
,
Fiazuddin
F
,
Strattan
EJ
,
Mo
X
,
Byrd
JC
,
Muthusamy
N
,
Awan
FT
.
Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia
.
Oncoimmunology
.
2016
:
5
(
10
):
e1226720
. https://doi.org/10.1080/2162402X.2016.1226720

43

André
P
,
Denis
C
,
Soulas
C
,
Bourbon-Caillet
C
,
Lopez
J
,
Arnoux
T
,
Bléry
M
,
Bonnafous
C
,
Gauthier
L
,
Morel
A
, et al.
Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells
.
Cell
.
2018
:
175
(
7
):
1731
1743.e13
. https://doi.org/10.1016/j.cell.2018.10.014

44

Kamiya
T
,
Seow
SV
,
Wong
D
,
Robinson
M
,
Campana
D
.
Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells
.
J Clin Invest
.
2019
:
129
(
5
):
2094
2106
. https://doi.org/10.1172/JCI123955

45

Lanier
LL
,
Chang
C
,
Phillips
JH
.
Human NKR-P1A. A disulfide-linked homodimer of the C-type lectin superfamily expressed by a subset of NK and T lymphocytes
.
J Immunol
.
1994
:
153
(
6
):
2417
2428
. https://doi.org/10.4049/jimmunol.153.6.2417

46

Tanaka
M
,
Fine
JH
,
Kirkham
CL
,
Aguilar
OA
,
Belcheva
A
,
Martin
A
,
Ketela
T
,
Moffat
J
,
Allan
DSJ
,
Carlyle
JR
.
The inhibitory NKR-P1B:Clr-b recognition axis facilitates detection of oncogenic transformation and cancer immunosurveillance
.
Cancer Res
.
2018
:
78
(
13
):
3589
3603
. https://doi.org/10.1158/0008-5472.CAN-17-1688

47

Vitale
C
,
Chiossone
L
,
Cantoni
C
,
Morreale
G
,
Cottalasso
F
,
Moretti
S
,
Pistorio
A
,
Haupt
R
,
Lanino
E
,
Dini
G
.
The corticosteroid-induced inhibitory effect on NK cell function reflects down-regulation and/or dysfunction of triggering receptors involved in natural cytotoxicity
.
Eur J Immunol
.
2004
:
34
(
11
):
3028
3038
. https://doi.org/10.1002/eji.200425418

48

Chiossone
L
,
Vitale
C
,
Cottalasso
F
,
Moretti
S
,
Azzarone
B
,
Moretta
L
,
Mingari
MC
.
Molecular analysis of the methylprednisolone-mediated inhibition of NK-cell function: evidence for different susceptibility of IL-2–versus IL-15–activated NK cells
.
Blood
.
2007
:
109
(
9
):
3767
3775
. https://doi.org/10.1182/blood-2006-07-037846

49

Gúma
M
,
Angulo
A
,
Vilches
C
,
Gómez-Lozano
N
,
Malats
N
,
López-Botet
M
.
Imprint of human cytomegalovirus infection on the NK cell receptor repertoire
.
Blood
.
2004
:
104
(
12
):
3664
3671
. https://doi.org/10.1182/blood-2004-05-2058

50

Muccio
L
,
Bertaina
A
,
Falco
M
,
Pende
D
,
Meazza
R
,
López-Botet
M
,
Moretta
L
,
Locatelli
F
,
Moretta
A
,
Della Chiesa
M
.
Analysis of memory-like natural killer cells in human cytomegalovirus-infected children undergoing αβ+ T and B cell-depleted hematopoietic stem cell transplantation for hematological malignancies
.
Haematologica
.
2016
:
101
(
3
):
371
381
. https://doi.org/10.3324/haematol.2015.134155

51

Ramírez-Ramírez
D
,
Padilla-Castañeda
S
,
Galán-Enríquez
CS
,
Vadillo
E
,
Prieto-Chávez
JL
,
Jiménez-Hernández
E
,
Vilchis-Ordóñez
A
,
Sandoval
A
,
Balandrán
JC
,
Pérez-Tapia
SM
.
CRTAM+ NK cells endowed with suppressor properties arise in leukemic bone marrow
.
J Leuk Biol
.
2019
:
105
(
5
):
999
1013
. https://doi.org/10.1002/JLB.MA0618-231R

Author notes

Conflict of interest statement. The authors declare no conflict of interest.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.