Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Clin Cancer Res. 2014 Oct 3;20(23):5986–5994. doi: 10.1158/1078-0432.CCR-14-0479

NK Cell Genotype and Phenotype at Diagnosis of Acute Lymphoblastic Leukemia Correlate to Post-induction Residual Disease

Erin M Sullivan 1, Sima Jeha 2,4, Guolian Kang 3, Cheng Cheng 3, Barbara Rooney 1, Martha Holladay 1, Rafijul Bari 1, Sarah Schell 1, MaCal Tuggle 1, Ching-Hon Pui 2,4, Wing Leung 1,4
PMCID: PMC4252745  NIHMSID: NIHMS633090  PMID: 25281696

Abstract

Purpose

Not all natural killer (NK) cells are equally cytotoxic against leukemia because of differences in receptor gene content and surface expression. We correlated NK cell genotype and phenotype at diagnosis of childhood acute lymphoblastic leukemia (ALL) with minimal residual disease (MRD) after induction chemotherapy.

Experimental Design

The NK cells and leukemia blasts of 244 patients were analyzed at diagnosis by killer-cell immunoglobulin-like receptor (KIR) typing and immunophenotyping. The results were correlated statistically to post-induction MRD status.

Results

The odds of being MRD positive in patients with KIR telomeric (Tel)-A/B genotype was 2.85 times the odds in those with Tel-A/A genotype (p=0.035). MRD positive patients were more likely to have KIR2DL5A (p=0.006) and expressed less activating receptor NKp46 and FASL on their NK cells (p=0.0074 and p=0.029, respectively). The odds of being MRD positive increased by 2.01-fold for every percentage increase in NK cells expressing KIR2DL1 in the presence of HLA-C2 ligand (p=0.034). The quantity of granzyme B inhibitor PI-9 in the leukemia blasts was greater in patients who were MRD positive (p=0.038). Collectively, five NK cell-related factors (Tel-B associated KIR2DL5A, NKp46, FASL, Granzyme B, and PI-9) are strongly associated with MRD positivity at the end of induction with 100% sensitivity and 80% specificity.

Conclusions

Our data support the hypothesis that NK cells with a strong effector phenotype in the setting of decreased leukemia resistance are associated with better leukemia control.

Keywords: NK cell factors, minimal residual disease, pediatric ALL

INTRODUCTION

The natural killer (NK) cell is a lymphocyte important in cancer control(1). A diverse repertoire of cell surface inhibitory and activating receptors dictates the NK cell’s response to both normal and abnormal cells. Once activated, the NK cell can lyse the target directly or secrete cytokines and chemokines that indirectly stimulate the host’s adaptive immune response(2).

NK cell cytotoxicity has been studied in hematological malignancies including acute myeloid leukemia (AML)(3, 4) and acute lymphoblastic leukemia (ALL)(5, 6). Additionally, many have examined the best use of NK cells to achieve maximum benefit in hematopoietic stem cell transplantation for AML or ALL(7, 8). Due to differences in gene content and variable surface receptor expression, not all NK cells are alike, however(7). We aimed to understand which NK cell-related factors are most relevant in childhood ALL therapy.

Early response to treatment is the most predictive marker for the risk of ALL relapse(9). It is because early response assessment by minimal residual disease (MRD) testing provides a precise and objective measurement of drug sensitivity of leukemia cells and the efficacy of treatment as well as host pharmacogenetics and immune surveillance(10). In this study, we prospectively investigated the relationship between features of the NK cells at the time of ALL diagnosis and MRD at the end of induction chemotherapy.

MATERIALS AND METHODS

Patients

Patients, 18 years old or younger, with newly diagnosed B- or T-lymphoblastic leukemia were enrolled on the institutional protocol Total Therapy Study XVI (TotalXVI). A total of 244 patients were enrolled from October, 2007 to October, 2011 at the time this laboratory research was performed. These studies were approved by the institutional review board. Written informed consent was obtained from the guardians, and assent from the patient, as appropriate.

Remission Induction Therapy

All patients received intrathecal chemotherapy with methotrexate, cytarabine, and hydrocortisone on day 1 and 15. Patients with Philadelphia chromosome, MLL rearrangement, hypodiploidy (<44 chromosomes), or WBC count ≥100 × 109/L at presentation received additional intrathecal treatment on days 8 and 22 and those with T cell ALL, t(1;19)/TCF3-PBX1, CNS 2 status (<5 WBC/μL CSF with blasts), CNS 3 status (≥5 WBC/μL of CSF with blasts or cranial nerve palsy) or traumatic lumbar puncture (>10 RBC with blasts) received additional intrathecal treatment on days 4 and 11. Induction treatment began with prednisone, vincristine, daunorubicin, and PEG asparaginase, followed by cyclophosphamide plus cytarabine and thioguanine. Dexamethasone was substituted for prednisone in patients with early T-cell precursor immunophenotype and mercaptopurine for thiopurine in those with thiopurine methyltransferase deficiency. An extra dose of PEG asparaginase was given if the day 15 MRD was ≥1% and fractionated cyclophosphamide at 300mg/m2 per dose every 12 hours for four doses was used instead of 1000mg/m2 for one dose if the day 15 MRD was ≥5%.

Assessment of response to induction therapy

Bone marrow aspirate for MRD determination was performed in all patients on day 38–42 of remission induction, when the absolute neutrophil count recovered to >300/μL, WBC >1000/μL, and platelets >50,000/μL. MRD was measured with flow cytometry or polymerase chain reaction (PCR) of immunoglobulin or T-cell receptor gene rearrangement, as described previously(10). MRD <0.01% was considered negative.

NK cell and leukemia blasts immunophenotyping

Peripheral whole blood (3 ml) was obtained at diagnosis for immunophenotyping by flow cytometry of NK cells and leukemic blasts. The following antibodies were used to enumerate NK cells and their surface expression: anti-KIR2DL1 (143211), anti-NTB-A (292811), anti-ULBP1 (170818), anti-ULBP2 (165903), anti-ULBP3 (166510), anti-MICA (159227), and anti-MICB (236511) were obtained from R&D Systems; anti-KIR3DL1 (DX9), anti-KIR2DL2/3 (CH-L), anti-CD11a (HI111), anti-NKG2D (1D11), anti-TRAIL (RIK-2), anti-DNAM (DX11), anti-2B4 (2–69), and anti-granzyme B (GB11) from BD Bioscience; anti-NKG2A (Z199), anti-NKp30 (Z25), anti-NKp44 (Z231), anti-NKp46 (BAB281), and anti-FAS (CIB2) from Beckman Coulter; and anti-FASL (14C2) from AbD Serotec. Blasts obtained at diagnosis were evaluated by anti-NTB-A (292811) from R&D Systems; anti-ICAM (84H10), anti-FAS (CIB2) and anti-CD48 (J4-57) from Beckman Coulter; anti-Nectin (R2.525) from BD Bioscience, anti-TRAIL-R1 (DJR1) and anti-TRAIL-R2 (DJR2-4) from Biolegend; anti-PVR (TX21) from MBL International; anti-HLA-ABCEFG (W6/32) from Dako; and anti-PI-9 (7D8) from AbD Serotec.

KIR genotyping

PCR amplification was performed with the Olerup SSP KIR genotyping kit (Qiagen). The following gene alleles were tested: 2DL1*001-010, 2DL2*001-005, 2DL3*001-007, 2DL5A*0010101-0010102 and 0050101-005010102, 2DL5B*00201010-004 and 00601-009, 2DS1*001-004, 2DS2*0010101-005, 2DS3*00101-004, 2DS4*00101010-00103, 003, 004, 006, 007 and 009, 2DS5*001-008, 3DL1*00101-002, 00401-009, 01501-044, 056 and 057, 3DL2*00101-021, 3DL3*00101-031, 3DS1*010-014, 045-049N and 055, 2DP1*00101-003, and 3DP1*001-006, 00301-00301 and 004-006. KIR2DL1 functional allele typing and KIR ligand typing were performed using a single nucleotide polymorphism (SNP) assay on the 7900 HT Sequence Detection System (Applied Biosystems) as described previously(11).

NK cell receptor transcripts

Quantification of NK cell transcripts was performed for the following: KIR2DS1-5, KIR2DL1-4, KIR3DL1-2, KIR3DS1, NKp30, NKp44, NKp46, and NKG2D. Real-time quantitative PCR (RQ-PCR) was performed by the 7900 HT Fast Real-Time PCR system. cDNA was obtained by performing reverse transcription on RNA using the Invitrogen Vilo kit (Invitrogen, Carlsbad, CA, USA).

PCR were performed with either 2.5 μL cDNA (from 12.5ng RNA), 2.5 μL of standards, or 2.5 μL of RNAse free water for the negative control. The master mix for all receptors, except KIR2DS3, included 12.5 μL of SYBR Green PCR master mix (Applied Biosystems), 2 μL of forward and reverse primers at a concentration of 5 μM, and 6 μL of RNAse free water. Standards were diluted in EB buffer from 105 copies down to 1 copy per 2.5μL. Cycling parameters were as follows: 50°C for 2 minutes, 95°C for 10 minutes, 40 cycles at 94°C for 30 seconds, 58°C for 30 seconds, and 72°C for 45 seconds, and 1 cycle at 72°C for 3 minutes. The dissociation steps were 95°C for 15 seconds, 60°C for 20 seconds, 95°C for 15 seconds.

The PCR for KIR2DS3 was performed with master mix containing 10μL of Fast SYBR Green and 3.5μL of RNAse and DNAse free water. Cycling parameters were as follows: 95°C for 20 seconds, 40 cycles at 95°C for 1 second and 60°C for 20 seconds, and a hold at 72°C for three minutes. The dissociation steps were the same as the setup for all other receptors.

Forward and reverse primers for receptors 2DL1-4, 3DL2, 2DS1-5, and 3DS1 are described previously(12). Forward and reverse primers for the remaining receptors were as follows: KIR3DL1 5′-CAAGCTCCAAATCTGGTAACCC-3′ and 5′-CCAACTGTGCGTATGTCACC-3′, NKp30 5′-CCCACTTGCTTCTTCCCGTTTCC-3′ and 5′-CACCACCAGCCGAGTCCCATTCC-3′, NKp44 5′-TCTCTAAGTCCGTCAGATTC-3′ and 5′-GATGGTAGATGGAGACTCAG-3′, NKp46 5′-ACGGGACTCCAGAAAGACCAT-3′ and 5′-CAGGCCCATCCGAAGGA-3′, and NKG2D 5′-GGCTCCATTCTCTCACCCA-3′ and 5′-TAAAGCTCGAGGCATAGAGTGC-3′.

Statistical analysis

We used the Fisher’s exact test and Wilcoxon rank sum test to compare categorical and continuous baseline variables between patients with positive or negative MRD at the end of induction chemotherapy, respectively. Univariate logistic regression was used to test associations between KIR genotype, NK cell receptor surface expression, leukemia blast characteristics, KIR haplotypes and MRD status, respectively. Wilcoxon rank sum test was used for comparison of the NK cell receptor mRNA transcript level between patients with positive or negative MRD. Receiver operating characteristic (ROC) curves, area under the curve (AUC) of the ROC, sensitivity, and specificity were calculated to determine levels of PI-9, FasL, Granzyme B, and NKp46 that best differentiate positive MRD vs. negative MRD using the maximum Youden index method implemented in R package pROC(13, 14). The smooth ROC curves were obtained using the method of maximum-likelihood fitting of univariate distributions (method = “fitdistr” in pROC). An MRD risk system based on the cutoffs of these four variables and the presence of KIR2DL5A was developed using logistic regression model. All the reported p-values are 2-sided and are considered significant if <0.05 because of exploratory nature of the study. Statistical analyses were performed with R-2.15.1(15).

RESULTS

Table 1 shows the presenting clinical and biological features of the 244 patients studied and the distribution of these features according to the MRD status at the end of induction. Not surprisingly, the MRD negative group was younger than the MRD positive group (p=0.0087).

Table 1.

Patient Characteristics

Characteristics All patients MRD Negative MRD Positive p-value
Number of patients 244 217 27
Age at Diagnosis (yrs) 0.0087
 Mean (SD) 7.14 (4.82) 6.86 (4.78) 9.36 (4.71)
 Median (Range) 5.44 (0.16–18.89) 5.26 (0.16–18.89) 9.71 (1.97–17.72)
Gender, N(%) 1
 Female 106 (43.44) 94 (43.32) 12 (44.44)
 Male 138 (56.56) 123 (56.68) 15 (55.56)
WBC at Diagnosis (103) 0.506
 Mean (SD) 39.41 (75.93) 38.59 (77.61) 46.01 (61.62)
 Median (Range) 10.15 (0.5–591.5) 10.1 (0.5–591.5) 20 (1.4–246.5)
Immunophenotype N(%) 0.78
 B-lineage 205 (84.02) 183 (84.33) 22 (81.48)
 T-lineage 39 (15.98) 34 (15.67) 5 (18.52)

NK cell Genotypes

Table 2 shows the proportion of patients positive for each KIR gene according to their MRD status. The frequency distribution of positivity in the entire cohort was no different than the general United States population(16). In a univariate logistic regression analysis, KIR2DL5A, KIR2DS1, and KIR2DS3 were statistically significant and were associated with increased odds of MRD by 3.05 to 4.5-fold. Notably, these three genes are found exclusively in the B haplotypes (Figure 1).

Table 2.

Correlation of KIR genotype with MRD

KIR gene % gene positive
OR (95%CI) p value
All patients Negative MRD Positive MRD
KIR2DL1 97.9 97.7 100 0(0-Inf) 0.99
KIR2DL2 53.7 54.7 44.4 0.66(0.25–1.76) 0.41
KIR2DL3 93.2 93 94.4 1.28(0.16–10.42) 0.82
KIR2DL5A 40 36.6 72.2 4.5(1.53–13.21) 0.006
KIR2DL5B 26.8 25.6 38.9 1.85(0.68–5.07) 0.23
KIR2DS1 31.6 29.1 55.6 3.05(1.14–8.18) 0.027
KIR2DS2 48.2 48.5 44.4 0.85(0.32–2.25) 0.74
KIR2DS3 24.2 21.5 50 3.65(1.35–9.85) 0.011
KIR2DS4*001 50 50 50 1(0.38–2.64) 1
KIR2DS4*003-9 74.7 74.4 77.8 1.2(0.38–3.85) 0.76
KIR2DS5 27.4 26.2 38.9 1.8 (0.66–4.92) 0.25
KIR3DL1 97.4 97.1 100 0(0-Inf) 0.99
KIR3DS1 30 27.9 50 2.58(0.97–6.9) 0.058
KIR2DP1 97.9 97.7 100 0(0-Inf) 0.99
KIR3DL1*004 25.4 25.7 22.2 0.82(0.26–2.64) 0.75

Figure 1.

Figure 1

Simplified maps of the A and B KIR haplotypes on chromosome 19q13.4. Cen-B is KIR2DL2 positive and KIR2DL3 negative in the centromeric motifs and Tel-B is KIR3DS1 positive and KIR3DL1 negative in the telomeric motifs.

The patients’ KIR genotypes were then categorized based on the presence of centromeric (Cen) or telomeric (Tel) A and B haplotype motifs as described previously(7). The odds of being MRD positive in patients with Tel A/B was 2.85 times than those with Tel A/A (95% CI 1.075–7.73, p = 0.035). No statistically significant difference was found between Cen A/A and Cen A/B. The effect of Cen B/B and Tel B/B was not examined because of the small number of patients (<15).

KIR mRNA and surface protein expression

Receptor transcripts and surface expression were quantified by RQ-PCR and flow cytometry, respectively. As there was no statistically significant difference between the MRD positive group and MRD negative group in the number of NK cells in the blood at diagnosis (p=0.12), the quantification of receptor transcripts was adjusted based on the amount of RNA (expressed as per μg of RNA, Table 3), rather than the amount of NK cells. Patients with positive MRD had a higher mRNA level of Tel B-associated KIR2DS1 (p=0.022) and a lower level of Tel A-associated KIR3DL1 (P=0.007) and KIR2DL1 transcripts (p=0.011) than did those with negative MRD (Table 3).

Table 3.

Correlation of NK cell receptor mRNA transcript level with MRD

Receptor Mean (SD) copies × 10 per μg of RNA
p value
All patients Negative MRD Positive MRD
KIR2DL1 44.6 (82.9) 48.9 (87.6) 16.4 (27.4) 0.011
KIR2DL2 19.1 (61.1) 20.9 (65.1) 7.6 (16.4) 0.21
KIR2DL3 51.3 (71.3) 54.3 (74.5) 31.5 (42) 0.11
KIR2DL4 26 (44.3) 24.5 (40.8) 35.9 (64.2) 0.19
KIR3DL1 75.8 (101.9) 83.6 (106.9) 23.8 (24.2) 0.007
KIR3DL2 1,388.3 (11,034.3) 1,547.4 (11,834.6) 335.8 (935.3) 0.14
KIR2DS1 8.2 (18.6) 7.8 (19.2) 11.4 (14.6) 0.022
KIR2DS2 37.5 (123.1) 35.5 (120.7) 50.4 (143.2) 0.36
KIR2DS3 4.8 (19.7) 5.2 (21) 2.5 (5.2) 0.96
KIR2DS4 137.2 (400.1) 137.8 (402.5) 133.3 (399.4) 0.3
KIR2DS5 4.3 (10.7) 4.4 (11.2) 3.9 (6.8) 0.63
KIR3DS1 19.8 (48) 17.4 (45.2) 36 (63.3) 0.074
NKp30 65.5 (87.1) 60.8 (68.6) 96.5 (165.5) 0.77
NKp44 1.1 (3.9) 1 (3.4) 1.8 (6.2) 0.29
NKp46 61.3 (80) 65.5 (84.3) 33.8 (32) 0.26
NKG2D 4,033.8 (17,096.3) 4,155.9 (18,124.6) 3,226.5 (7,698.3) 0.36

The association of MRD with surface expression of KIR2DL1, KIR2DL2/3, and KIR3DL1 as enumerated by flow cytometry was analyzed in the context of the presence of their corresponding ligand. The odds of being MRD positive in a patient with HLAC2 ligand increased by 2.01-fold (95% CI 1.05–3.85) for every percentage increase in NK cells expressing KIR2DL1 on their surface (p = 0.034). There were no significant association between MRD and receptor-ligand pairs for KIR2DL2, KIR2DL3 and KIR3DL1.

Other NK cell receptor surface expression

The proportion of NK cells expressing NKG2A, NKp46, and FAS receptor were significantly associated with the MRD status. The NK cells from the MRD positive patients had lower frequency of expression of NKG2A (mean ± SD, 27.1% ± 16.1% vs. 38.3 ± 14.3%, p = 0.0012) and NKp46 (37.2% ± 37% vs. 56.6% ± 29%, p=0.0074), but higher expression of FAS (44.8% ± 43.2% vs. 23.8% ± 31.8%, p=0.032) than NK cells from MRD negative patients. In terms of surface density, the mean florescence intensity of FASL was lower in the MRD positive group than the MRD negative group (348 ± 201.3 vs. 543.1 ± 358.7, p=0.029).

Leukemia Blast Characteristics

Among the 13 biomarkers, ICAM, PI-9, and NTBA were statistically associated with MRD status. The leukemia blasts from the MRD positive patients had a greater frequency of expression of ICAM (mean ± SD, 34.9% ±30.2% vs. 22.5% ±24.5%, p=0.035) and PI-9 (mean ± SD, 63.3% ± 35% vs. 45.6% ±35.2%, p=0.027) than blasts in the MRD negative group. Further, the amount of PI-9 in the MRD positive group was greater than that in the MRD negative group (MFI mean ± SD, 996 ± 677.8 vs. 691.6 ± 527.5, p=0.038). The amount of NTB-A expressed on the blasts was also greater in the MRD positive group (MFI mean ± SD, 235.9 ±186.5 vs. 184.1 ± 79.4, p=0.04). There was no statistically significant difference between the MRD positive group and MRD negative group in the expression of HLA-Class I on the leukemia blasts (MFI mean±SD, 20.5±19.7 vs. 18.8±17.4 × 103, p=0.64).

Composite Model

Among the significant results of immunophenotyping and KIR genotyping, five NK-related parameters were revealed by receiver operating characteristics (ROC) analyses to be highly correlated collectively with the risk of MRD, including PI-9, FasL, Granzyme B, NKp46 and KIR2DL5A. The probability of positive MRD ranged from as low as 3.32 ×10−9 % to as high as 81.6% (Table 4), using cutoffs which were established statistically from individual ROC (>650, 415, 4070, and 30 for the first four predictors, respectively, and the presence of KIR2DL5A). The five-parameter composite model had a sensitivity of 100% and specificity of 80% (Figure 2); the AUC of its ROC curve improved to 0.93 when compared with those of individual factors (all AUC <0.67).

Table 4.

Probability of positive MRD post-induction based on five NK-related parameters.

Risk Category Group Blast PI-9 MFI NK FasL MFI NK Granzyme B MFI NK NKp46 % NK KIR2DL5A genotype Probability MRD positive (%)
Low 1 <=650 >415 > 4070 > 30 3.32 × 10−9
2 <=650 > 415 > 4070 > 30 + 1.34 × 10−8
3 <=650 <=415 > 4070 > 30 1.87 × 10−8
4 > 650 > 415 > 4070 > 30 2.36 × 10−8
5 <=650 > 415 > 4070 <=30 3.72 × 10−8
6 <=650 <=415 > 4070 > 30 + 7.57 × 10−8
7 > 650 > 415 > 4070 > 30 + 9.57 × 10−8
8 > 650 <=415 > 4070 > 30 1.33 × 10−7
9 <=650 > 415 > 4070 <=30 + 1.51 × 10−7
10 <=650 <=415 > 4070 <=30 2.09 × 10−7
11 > 650 > 415 > 4070 <=30 2.64 × 10−7
12 > 650 <=415 > 4070 > 30 + 5.38 × 10−7
13 <=650 <=415 > 4070 <=30 + 8.47 × 10−7
14 > 650 > 415 > 4070 <=30 + 1.07 × 10−6
15 > 650 <=415 > 4070 <=30 1.49 × 10−6
16 > 650 <=415 > 4070 <=30 + 6.03 × 10−6
Intermediate 17 <=650 > 415 <=4070 > 30 0.2
18 <=650 > 415 <=4070 > 30 + 1
19 <=650 <=415 <=4070 > 30 1.4
20 > 650 > 415 <=4070 > 30 1.7
21 <=650 > 415 <=4070 <=30 2.7
22 <=650 <=415 <=4070 > 30 + 5.3
23 > 650 > 415 <=4070 > 30 + 6.6
24 > 650 <=415 <=4070 > 30 8.9
25 <=650 > 415 <=4070 <=30 + 9.9
High 26 <=650 <=415 <=4070 <=30 13.3
27 > 650 > 415 <=4070 <=30 16.2
28 > 650 <=415 <=4070 > 30 + 28.3
29 <=650 <=415 <=4070 <=30 + 38.3
30 > 650 > 415 <=4070 <=30 + 44
31 > 650 <=415 <=4070 <=30 52.2
32 > 650 <=415 <=4070 <=30 + 81.6

Figure 2.

Figure 2

The five NK cell related factors were analyzed either singly or as composite in correlation with post-induction MRD. The composite model has a 100% sensitivity (or 100% true positivity) and an 80% specificity (or 20% false positivity).

DISCUSSION

The activity of NK cells is regulated by the engagement of their inhibitory and activating surface receptors with ligands on the leukemia lymphoblasts. Cytotoxicity is achieved through perforin/granzymes or FasL(2, 17). Herein, we presented our comprehensive evaluation of these molecules in NK cells and target leukemia cells, and identified five NK-related parameters that correlated strongly with MRD after induction chemotherapy for childhood ALL.

KIR diversity is generated through haplotype gene content on chromosome 19q13.4, allele polymorphism, and stochastic surface expression(18, 19). In our study, the presence of genes KIR2DL5A, KIR2DS1, and KIR2DS3 were significantly associated with a positive MRD. Interestingly, these three genes are exclusively found in group-B KIR haplotypes, suggesting that B haplotypes are associated with a poor induction response in pediatric ALL. When we compared the haplotypes A/A versus A/B directly, the telomeric rather than the centromeric B motifs appeared to be the key determinant, as the odds of being MRD positive with Tel-A/B genotypes was significantly higher than that of Tel-A/A, whereas no difference was observed between Cen-A/B and Cen-A/A. In contrast to our study, some investigators have found that the group-B KIR haplotypes were associated with improved overall survival and event-free survival, as well as lower transplant-related mortality(20, 21). However, others corroborate our findings of an A haplotype benefit and a B haplotype disadvantage(22, 23). The A haplotypes were associated with a complete molecular response in chronic myeloid leukemia(24) and an improved disease free survival in patients receiving a haploidentical T-cell depleted stem cell transplant for leukemia. The risk of relapse in the latter study increased with the number of activating donor KIR genes present(25). Because B haplotypes typically contain larger numbers of activating KIR genes than the A haplotypes (which contain only two activating genes KIR2DL4 and KIR2DS4 that are often disrupted by mutations rendering them non-functional)(2628), these data collectively suggest that the risk of relapse might be dependent on the activating gene content in the B haplotypes. In this regard, NK cells possessing Tel-B associated KIR2DS1 have been shown to be hypo-responsive because of tolerance induced by homozygous C2 ligands(29, 30).

To further dissect the role of activating KIRs from that of inhibitory KIRs in MRD, we directly measured the patients’ KIR repertoire by RQ-PCR. This approach had two advantages. First, the RQ-PCR allowed us to analyze all 15 family members of KIRs individually, as monoclonal antibodies for flow cytometry are either not available or cannot discern the activating versus inhibitory KIRs. Second, previous studies have shown that genotypes do not always correlate with phenotypes(31, 32). In our cohort, the MRD positive group had more mRNA transcripts of the B-haplotypes associated KIR2DS1 activating receptor than the MRD negative group, collaborating with our genotype findings of detrimental Tel-B effect. By contrast, the MRD negative group expressed more mRNA of inhibitory receptors KIR2DL1 and A-haplotypes associated inhibitory KIR3DL1, supporting the hypothesis of the importance of inhibitory KIR acquisition during NK cell licensing for missing-self recognition(3335). The licensing or arming model proposes that inhibitory receptor and MHC ligand interactions educate the NK cells to kill when it encounters a target cell missing the cognate ligand(3639). The greater the number of inhibitory receptors on NK cells, the stronger the cytotoxic responsiveness(40, 41). The absence of these inhibitory receptors, even in the presence of activating receptors, leaves the NK cell hypo-responsive. Alternatively, our Tel-B genotype and RQ-PCR data also supports the disarming model, which states that NK cells exhibiting more stimulatory signal become dampened overtime(42, 43). In our patients, those with a poor response to induction chemotherapy are typified by the Tel-B genotypes with high level of expression of activating KIR2DS1 and low level of expression of inhibitory KIR2DL1 and KIR3DL1, a phenotype found in immature NK cells and unlicensed NK cells(44, 45). In fact, KIR2DL5A is the most significant risk factor for MRD. Although the function and ligand of KIR2DL5A are unknown, this KIR gene is almost in complete linkage disequilibrium with the absence of Tel-KIR3DL1and the presence of Tel-KIR2DS1, KIR2DS3, KIR2DS5, and KIR3DS1; NK cells of these genotypes (more activating and less inhibitory gene content) would be predictably hypo-responsive based on the arming and disarming model(42, 43).

In addition to KIRs, other NK cell receptors and effector molecules are crucial determinants of antileukemia response(2, 7, 17, 46). A NCRdull phenotype, for example, has been associated with poor leukemia control in adult AML(3, 4), in line with our data showing that MRD was associated with low expression of NKp46. Furthermore, the amount of FasL in the NK cells of the MRD positive group was smaller than that of the MRD negative group; FasL is essential to induce the death pathway in the target cells(47).

The leukemia cell-intrinsic factors may also play a role in NK-cell escape. In our patients, MRD positivity was associated with the presence of C2-ligand and the abundance of intracellular PI-9. While C2 may inhibit KIR2DL1, PI-9 can resist granzyme B(48) or FasL mediated cytotoxicity, rendering both cytotoxic pathways ineffective(49).

Post-induction MRD has been established to be one of the most important prognostic markers for childhood ALL and MRD-based risk-adaptive continuation regimens have been shown to improve patient outcomes(50). Herein, we identified five NK-related factors that collectively correlated strongly with post-induction MRD. For instance, patients in groups 1–16 as listed in Table 4 had a <1×10−5% risk of positive MRD; these patients would be expected to have an excellent outcome after treatment with TotalXVI regimen. By contrast, patients in groups 17–25 had an intermediate risk ranging from 0.2% to 10%, whereas those in the high risk groups had a risk ranging from 13.3% to 81.6%. The composite model had favorable operating characteristics (100% sensitivity and 80% specificity, corresponding with a 100% negative predictive value), thus providing strong support to our biologic hypothesis that NK cells with a strong effector phenotype in the setting of decrease leukemia resistance are associated with better leukemia control.

TRANSLATIONAL RELEVANCE.

Post-induction minimal residual disease (MRD) in pediatric acute lymphoblastic leukemia has been established to be one of the most important prognostic markers. MRD based risk-adaptive continuation chemotherapy regimens have been shown to improve patient outcomes. Natural killer (NK) cells are lymphocytes important in leukemia control. However, not all NK cells are alike. We identified five NK-related factors that collectively correlate strongly with postinduction MRD. These factors include Fas Ligand, Granzyme B, NKp46, and KIR2DL5A genotype in the NK cell and PI-9 in the leukemia blast. This knowledge allows clinicians to analyze NK cell and leukemia blast features at the time of diagnosis and make predictions about MRD at the end of induction and therefore overall prognosis.

Acknowledgments

Financial support: The manuscript was supported in part by research grant No. CA-21765 from the National Institute of Health and by the Assisi Foundation of Memphis.

Cheng Cheng - Research funding from NIH and Alpha Tau Pharmaceuticals.

Ching-Hon Pui - Research funding from NCI CA36401 and GM92666

Footnotes

Conflicts of Interest Disclosures:

Erin Sullivan, Sima Jeha, Guolian Kang, Barbara Rooney, Martha Holladay, Rafijul Bari, Sarah Schell, MaCal Tuggle, and Wing Leung have no conflicts of interest to disclose.

References

  • 1.Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet. 2000;356:1795–9. doi: 10.1016/S0140-6736(00)03231-1. [DOI] [PubMed] [Google Scholar]
  • 2.Caligiuri MA. Human natural killer cells. Blood. 2008;112:461–9. doi: 10.1182/blood-2007-09-077438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Costello RT, Sivori S, Marcenaro E, Lafage-Pochitaloff M, Mozziconacci MJ, Reviron D, et al. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood. 2002;99:3661–7. doi: 10.1182/blood.v99.10.3661. [DOI] [PubMed] [Google Scholar]
  • 4.Fauriat C, Just-Landi S, Mallet F, Arnoulet C, Sainty D, Olive D, et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood. 2007;109:323–30. doi: 10.1182/blood-2005-08-027979. [DOI] [PubMed] [Google Scholar]
  • 5.Nasrallah AG, Miale TD. Decreased natural killer cell activity in children with untreated acute leukemia. Cancer Res. 1983;43:5580–5. [PubMed] [Google Scholar]
  • 6.Dickinson AM, Proctor SJ, Jacobs E, Reid MM, Walker W, Craft AW, et al. Natural killer cell activity in childhood acute lymphoblastic leukaemia in remission. Br J Haematol. 1985;59:45–53. doi: 10.1111/j.1365-2141.1985.tb02962.x. [DOI] [PubMed] [Google Scholar]
  • 7.Leung W. Use of NK cell activity in cure by transplant. Br J Haematol. 2011;155:14–29. doi: 10.1111/j.1365-2141.2011.08823.x. [DOI] [PubMed] [Google Scholar]
  • 8.Velardi A, Ruggeri L, Mancusi A. Killer-cell immunoglobulin-like receptors reactivity and outcome of stem cell transplant. Curr Opin Hematol. 2012;19:319–23. doi: 10.1097/MOH.0b013e32835423c3. [DOI] [PubMed] [Google Scholar]
  • 9.Inaba H, Greaves M, Mullighan CG. Acute lymphoblastic leukaemia. Lancet. 2013;381:1943–55. doi: 10.1016/S0140-6736(12)62187-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Campana D. Minimal residual disease in acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program. 2010;2010:7–12. doi: 10.1182/asheducation-2010.1.7. [DOI] [PubMed] [Google Scholar]
  • 11.Bari R, Rujkijyanont P, Sullivan E, Kang G, Turner V, Gan K, et al. Effect of Donor KIR2DL1 Allelic Polymorphism on the Outcome of Pediatric Allogeneic Hematopoietic Stem-Cell Transplantation. Journal of Clinical Oncology. 2013 doi: 10.1200/JCO.2012.47.4007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Thompson A, van der Slik AR, Koning F, van Bergen J. An improved RT-PCR method for the detection of killer-cell immunoglobulin-like receptor (KIR) transcripts. Immunogenetics. 2006;58:865–72. doi: 10.1007/s00251-006-0163-9. [DOI] [PubMed] [Google Scholar]
  • 13.Youden WJ. Index for rating diagnostic tests. Cancer. 1950;3:32–5. doi: 10.1002/1097-0142(1950)3:1<32::aid-cncr2820030106>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 14.Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez JC, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics. 2011;12:77. doi: 10.1186/1471-2105-12-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.R Development Core Team. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2010. [Google Scholar]
  • 16.Gonzalez-Galarza FF, Christmas S, Middleton D, Jones AR. Allele frequency net: a database and online repository for immune gene frequencies in worldwide populations. Nucleic Acids Res. 2011;39:D913–9. doi: 10.1093/nar/gkq1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Screpanti V, Wallin RP, Grandien A, Ljunggren HG. Impact of FASL-induced apoptosis in the elimination of tumor cells by NK cells. Mol Immunol. 2005;42:495–9. doi: 10.1016/j.molimm.2004.07.033. [DOI] [PubMed] [Google Scholar]
  • 18.Bari R, Bell T, Leung WH, Vong QP, Chan WK, Das Gupta N, et al. Significant functional heterogeneity among KIR2DL1 alleles and a pivotal role of arginine 245. Blood. 2009;114:5182–90. doi: 10.1182/blood-2009-07-231977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Parham P. MHC class I molecules and KIRs in human history, health and survival. Nat Rev Immunol. 2005;5:201–14. doi: 10.1038/nri1570. [DOI] [PubMed] [Google Scholar]
  • 20.Symons HJ, Leffell MS, Rossiter ND, Zahurak M, Jones RJ, Fuchs EJ. Improved survival with inhibitory killer immunoglobulin receptor (KIR) gene mismatches and KIR haplotype B donors after nonmyeloablative, HLA-haploidentical bone marrow transplantation. Biol Blood Marrow Transplant. 2010;16:533–42. doi: 10.1016/j.bbmt.2009.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cooley S, Weisdorf DJ, Guethlein LA, Klein JP, Wang T, Le CT, et al. Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia. Blood. 2010;116:2411–9. doi: 10.1182/blood-2010-05-283051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Verheyden S, Bernier M, Demanet C. Identification of natural killer cell receptor phenotypes associated with leukemia. Leukemia. 2004;18:2002–7. doi: 10.1038/sj.leu.2403525. [DOI] [PubMed] [Google Scholar]
  • 23.McQueen KL, Dorighi KM, Guethlein LA, Wong R, Sanjanwala B, Parham P. Donor-recipient combinations of group A and B KIR haplotypes and HLA class I ligand affect the outcome of HLA-matched, sibling donor hematopoietic cell transplantation. Hum Immunol. 2007;68:309–23. doi: 10.1016/j.humimm.2007.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.La Nasa G, Caocci G, Littera R, Atzeni S, Vacca A, Mulas O, et al. Homozygosity for killer immunoglobin-like receptor haplotype A predicts complete molecular response to treatment with tyrosine kinase inhibitors in chronic myeloid leukemia patients. Exp Hematol. 2013;41:424–31. doi: 10.1016/j.exphem.2013.01.008. [DOI] [PubMed] [Google Scholar]
  • 25.Kroger N, Binder T, Zabelina T, Wolschke C, Schieder H, Renges H, et al. Low number of donor activating killer immunoglobulin-like receptors (KIR) genes but not KIR-ligand mismatch prevents relapse and improves disease-free survival in leukemia patients after in vivo T-cell depleted unrelated stem cell transplantation. Transplantation. 2006;82:1024–30. doi: 10.1097/01.tp.0000235859.24513.43. [DOI] [PubMed] [Google Scholar]
  • 26.Parham P. Immunogenetics of killer-cell immunoglobulin-like receptors. Tissue Antigens. 2003;62:194–200. doi: 10.1034/j.1399-0039.2003.00126.x. [DOI] [PubMed] [Google Scholar]
  • 27.Goodridge JP, Lathbury LJ, Steiner NK, Shulse CN, Pullikotil P, Seidah NG, et al. Three common alleles of KIR2DL4 (CD158d) encode constitutively expressed, inducible and secreted receptors in NK cells. Eur J Immunol. 2007;37:199–211. doi: 10.1002/eji.200636316. [DOI] [PubMed] [Google Scholar]
  • 28.Middleton D, Gonzalez A, Gilmore PM. Studies on the expression of the deleted KIR2DS4*003 gene product and distribution of KIR2DS4 deleted and nondeleted versions in different populations. Hum Immunol. 2007;68:128–34. doi: 10.1016/j.humimm.2006.12.007. [DOI] [PubMed] [Google Scholar]
  • 29.Chewning JH, Gudme CN, Hsu KC, Selvakumar A, Dupont B. KIR2DS1-positive NK cells mediate alloresponse against the C2 HLA-KIR ligand group in vitro. J Immunol. 2007;179:854–68. doi: 10.4049/jimmunol.179.2.854. [DOI] [PubMed] [Google Scholar]
  • 30.Pittari G, Liu XR, Selvakumar A, Zhao Z, Merino E, Huse M, et al. NK cell tolerance of self-specific activating receptor KIR2DS1 in individuals with cognate HLA-C2 ligand. J Immunol. 2013;190:4650–60. doi: 10.4049/jimmunol.1202120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Leung W, Iyengar R, Triplett B, Turner V, Behm FG, Holladay MS, et al. Comparison of killer Ig-like receptor genotyping and phenotyping for selection of allogeneic blood stem cell donors. J Immunol. 2005;174:6540–5. doi: 10.4049/jimmunol.174.10.6540. [DOI] [PubMed] [Google Scholar]
  • 32.McErlean C, Gonzalez AA, Cunningham R, Meenagh A, Shovlin T, Middleton D. Differential RNA expression of KIR alleles. Immunogenetics. 2010;62:431–40. doi: 10.1007/s00251-010-0449-9. [DOI] [PubMed] [Google Scholar]
  • 33.Fernandez NC, Treiner E, Vance RE, Jamieson AM, Lemieux S, Raulet DH. A subset of natural killer cells achieves self-tolerance without expressing inhibitory receptors specific for self-MHC molecules. Blood. 2005;105:4416–23. doi: 10.1182/blood-2004-08-3156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Anfossi N, Andre P, Guia S, Falk CS, Roetynck S, Stewart CA, et al. Human NK cell education by inhibitory receptors for MHC class I. Immunity. 2006;25:331–42. doi: 10.1016/j.immuni.2006.06.013. [DOI] [PubMed] [Google Scholar]
  • 35.Hasenkamp J, Borgerding A, Uhrberg M, Falk C, Chapuy B, Wulf G, et al. Self-tolerance of human natural killer cells lacking self-HLA-specific inhibitory receptors. Scand J Immunol. 2008;67:218–29. doi: 10.1111/j.1365-3083.2007.02058.x. [DOI] [PubMed] [Google Scholar]
  • 36.Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song YJ, Yang L, et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature. 2005;436:709–13. doi: 10.1038/nature03847. [DOI] [PubMed] [Google Scholar]
  • 37.Yokoyama WM, Kim S. How do natural killer cells find self to achieve tolerance? Immunity. 2006;24:249–57. doi: 10.1016/j.immuni.2006.03.006. [DOI] [PubMed] [Google Scholar]
  • 38.Yokoyama WM, Kim S. Licensing of natural killer cells by self-major histocompatibility complex class I. Immunol Rev. 2006;214:143–54. doi: 10.1111/j.1600-065X.2006.00458.x. [DOI] [PubMed] [Google Scholar]
  • 39.Karre K. NK cells, MHC class I molecules and the missing self. Scand J Immunol. 2002;55:221–8. doi: 10.1046/j.1365-3083.2002.01053.x. [DOI] [PubMed] [Google Scholar]
  • 40.Yu J, Heller G, Chewning J, Kim S, Yokoyama WM, Hsu KC. Hierarchy of the human natural killer cell response is determined by class and quantity of inhibitory receptors for self-HLA-B and HLA-C ligands. J Immunol. 2007;179:5977–89. doi: 10.4049/jimmunol.179.9.5977. [DOI] [PubMed] [Google Scholar]
  • 41.Joncker NT, Fernandez NC, Treiner E, Vivier E, Raulet DH. NK cell responsiveness is tuned commensurate with the number of inhibitory receptors for self-MHC class I: the rheostat model. J Immunol. 2009;182:4572–80. doi: 10.4049/jimmunol.0803900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Raulet DH, Vance RE. Self-tolerance of natural killer cells. Nat Rev Immunol. 2006;6:520–31. doi: 10.1038/nri1863. [DOI] [PubMed] [Google Scholar]
  • 43.Joncker NT, Raulet DH. Regulation of NK cell responsiveness to achieve self-tolerance and maximal responses to diseased target cells. Immunol Rev. 2008;224:85–97. doi: 10.1111/j.1600-065X.2008.00658.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cooley S, Xiao F, Pitt M, Gleason M, McCullar V, Bergemann TL, et al. A subpopulation of human peripheral blood NK cells that lacks inhibitory receptors for self-MHC is developmentally immature. Blood. 2007;110:578–86. doi: 10.1182/blood-2006-07-036228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Montaldo E, Del Zotto G, Della Chiesa M, Mingari MC, Moretta A, De Maria A, et al. Human NK cell receptors/markers: A tool to analyze NK cell development, subsets and function. Cytometry A. 2013 doi: 10.1002/cyto.a.22302. [DOI] [PubMed] [Google Scholar]
  • 46.Leung W. Infusions of Allogeneic Natural Killer Cells as Cancer Therapy. Clin Cancer Res. 2014;20:3390–400. doi: 10.1158/1078-0432.CCR-13-1766. [DOI] [PubMed] [Google Scholar]
  • 47.Screpanti V, Wallin RP, Ljunggren HG, Grandien A. A central role for death receptor-mediated apoptosis in the rejection of tumors by NK cells. J Immunol. 2001;167:2068–73. doi: 10.4049/jimmunol.167.4.2068. [DOI] [PubMed] [Google Scholar]
  • 48.Classen CF, Ushmorov A, Bird P, Debatin KM. The granzyme B inhibitor PI-9 is differentially expressed in all main subtypes of pediatric acute lymphoblastic leukemias. Haematologica. 2004;89:1314–21. [PubMed] [Google Scholar]
  • 49.Cunningham TD, Jiang X, Shapiro DJ. Expression of high levels of human proteinase inhibitor 9 blocks both perforin/granzyme and Fas/Fas ligand-mediated cytotoxicity. Cell Immunol. 2007;245:32–41. doi: 10.1016/j.cellimm.2007.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pui CH, Campana D, Pei D, Bowman WP, Sandlund JT, Kaste SC, et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med. 2009;360:2730–41. doi: 10.1056/NEJMoa0900386. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES