Next Article in Journal
Targeting the NLRP3 Inflammasome as a New Therapeutic Option for Overcoming Cancer
Next Article in Special Issue
Defucosylation of Tumor-Specific Humanized Anti-MUC1 Monoclonal Antibody Enhances NK Cell-Mediated Anti-Tumor Cell Cytotoxicity
Previous Article in Journal
Recent Advances in Implantation-Based Genetic Modeling of Biliary Carcinogenesis in Mice
Previous Article in Special Issue
Hypoxia-Driven HIF-1α Activation Reprograms Pre-Activated NK Cells towards Highly Potent Effector Phenotypes via ERK/STAT3 Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Expression of NK Cell Receptor Ligands on Leukemic Cells Is Associated with the Outcome of Childhood Acute Leukemia

by
María Victoria Martínez-Sánchez
1,
José Luis Fuster
2,
José Antonio Campillo
1,
Ana María Galera
2,
Mar Bermúdez-Cortés
2,
María Esther Llinares
2,
Eduardo Ramos-Elbal
2,
Juan Francisco Pascual-Gázquez
2,
Ana María Fita
2,
Helios Martínez-Banaclocha
1,
José Antonio Galián
1,
Lourdes Gimeno
1,3,
Manuel Muro
1 and
Alfredo Minguela
1,*
1
Immunology Service, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain
2
Pediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca and Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain
3
Human Anatomy Department, University of Murcia (UM), 30100 Murcia, Spain
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(10), 2294; https://doi.org/10.3390/cancers13102294
Submission received: 24 March 2021 / Revised: 21 April 2021 / Accepted: 6 May 2021 / Published: 11 May 2021
(This article belongs to the Special Issue Natural Killer Cells in Cancers)

Abstract

:

Simple Summary

Natural killer cells (NK cells) of the innate immune system are suspected of playing an important role in eliminating residual leukemia cells during maintenance chemotherapy given to children with acute lymphoblastic leukemia for about two years. This study analyzes the expression of ligands for the receptors that regulate the function of NK cells on leukemic cells of more than one hundred children with acute lymphoid and myeloid leukemia. Our results show that the loss of expression of some molecules involved in the activation of NK cells is associated with poorer survival. In addition, a genetic combination of molecules that interact to regulate NK cell function seems to be associated with a higher relapse rate during/after chemotherapy and shorter patient survival. Children who carry this genetic combination are refractory to current chemotherapy treatments, and stem cell transplantation does not seem to contribute to their cure either, and therefore, they should be considered as candidates for alternative biological therapies that might offer better results.

Abstract

Acute leukemia is the most common malignancy in children. Most patients are cured, but refractory/relapsed AML and ALL are the first cause of death from malignancy in children. Maintenance chemotherapy in ALL has improved survival by inducing leukemic cell apoptosis, but immune surveillance effectors such as NK cells might also contribute. The outcome of B-ALL (n = 70), T-ALL (n = 16), and AML (n = 16) pediatric patients was evaluated according to leukemic cell expression of ligands for activating and inhibiting receptors that regulate NK cell functioning. Increased expression of ULBP-1, a ligand for NKG2D, but not that of CD112 or CD155, ligands for DNAM-1, was associated with poorer 5-year event-free survival (5y-EFS, 77.6% vs. 94.9%, p < 0.03). Reduced expression of HLA-C on leukemic cells in patients with the KIR2DL1/HLA-C*04 interaction was associated with a higher rate of relapse (17.6% vs. 4.4%, p = 0.035) and lower 5y-EFS (70.6% vs. 92.6%, p < 0.002). KIR2DL1/HLA-C*04 interaction was an independent predictive factor of events (HR = 4.795, p < 0.005) or death (HR = 6.731, p < 0.005) and might provide additional information to the current risk stratification. Children who carry the KIR2DL1/HLA-C*04 interaction were refractory to current chemotherapy treatments, including allogeneic stem cell transplantation; therefore, they should be considered as candidates for alternative biological therapies that might offer better results.

Graphical Abstract

1. Introduction

Acute leukemia is the most common malignancy in children and is characterized by the accumulation of leukocyte progenitors in the bone marrow (BM) [1]. In the last two decades, great progress has been made in the biological understanding of the disease, thus promoting molecular diagnosis, patient risk stratification, therapy personalization, early evaluation of treatment response, and substantial improvement in disease management and clinical outcome. Nonetheless, current treatment results of childhood acute myeloblastic leukemia (AML) are considered sub-optimal and new drugs are being evaluated [1]. In contrast, most children with acute lymphoblastic leukemia (ALL) are cured with current multi-agent chemotherapy regimens, including in most patients a maintenance phase of less intensive chemotherapy for a prolonged period of time, which is critical for ensuring the cure of the disease [2]. Despite these encouraging advances, the outcome of patients with refractory/relapsed AML and ALL is dismal, and these types of leukemia still are the most common cause of death from malignancy in children [3]. The ALL therapeutic landscape has changed dramatically with the incorporation of novel immunotherapies, such as the bispecific T-cell engager (BiTE) that links CD19+ B-cells with CD3+ T cells, the calicheamicin-conjugated anti-CD22 monoclonal antibody, and multiple CD19-targeted chimeric antigen receptor (CAR) T cells [4,5,6,7]. Natural killer (NK) cell therapies are also offering new opportunities to patients with refractory AML and ALL, either through hematopoietic stem cell transplantation (HSCT) with donors selected to maximize NK-cell alloreactivity [8,9] or through ex vivo activation of patients’ own NK cells or allogeneic NK cells [10,11]. To improve the efficacy of these treatments [12], advances in the knowledge of the interactions that govern NK cell function are needed, particularly those interactions promoted by molecules expressed on leukemic cells that could define the type and potency of NK cell antitumor response.
Antileukemic NK cell response is regulated by the integration of signals from activating FcγRIII, NKG2D, DNAM-1, NKp30, NKp44, or NKp46 receptors [13] and inhibitory NKG2A or killer-cell immunoglobulin-like receptors (KIR) when interacting with their specific ligands on the transformed cells. The balance between positive and negative signals allows NK cells to differentiate healthy tissues from tissues expressing damage/danger ligands for activating receptors or from tissues with loss of ligands for inhibitory receptors such as human leukocyte antigens class I (HLA-I), also known as missing-self [14,15]. The loss of HLA-I expression to escape the cytolytic activity of T lymphocytes is a frequent event in cancer [16], and it is in these circumstances that the antitumor function of NK cells becomes particularly relevant [17]. However, NK cells are also able to sense alterations in the peptidome presented by specific HLA allotypes that may occur during viral infections or malignant transformations [18,19]. For this reason, the patient’s genetic profile, particularly the profile of the highly polymorphic KIR/HLA-I interactions, can condition the immune response, and thus, the appearance and progression of malignant diseases.
To better understand NK cell functioning, we must take into account that during their development, the recognition of their own HLA-I by NKG2A and inhibitory KIR (iKIR) receptors promotes their maturation and functional competence, educating them in a process called “licensing”. This increases the expression of the DNAM-1 activating receptor and the stability of NK/tumor cell synapse [20]. On the contrary, the lack of self-recognition through inhibitory receptors leads to incomplete maturation and hyporesponsive NK cells [21,22,23]. Inhibitory licensing interactions include KIR2DL1/C2-epitope (HLA-C allotypes with Lys80 in alpha-1 helix), KIR2DL2-3/C1-epitope (HLA-C allotypes with Asn80 and the rare allotypes HLA-B*46:01 and HLA-B*73:01) [18,24], KIR3DL1/Bw4-epitope (HLA-A and HLA-B allotypes with Bw4 epitope, either with Thr80 -80T- or Ile80 -80I-) [25], KIR3DL2/HLA-A*03 and A*11 allotypes [26], and NKG2A/HLA-E [27]. KIR2DL2-3 can also interact with C2-epitope, although with lower affinity [18].
Interactions for activating KIRs (aKIR) have been reported for KIR2DS1/HLA-C2 [28,29], KIR2DS2/C1 [30], KIR2DS2/HLA-A*11 [31], KIR2DS2/non-HLA ligands expressed in cancer [32], KIR2DS4/HLA-A*11, C*01:02, C*02:02, C*04:01, C*05:01, C*14:02, and C*16:01 [33]. KIR3DS1 ligands have remained indefinite until recently, and they are represented by the open conformation of the non-classical HLA-F molecule and by the HLA-B*51 (an HLA-B Bw4 80I allotype) [18,34,35]. KIR2DS5*006 (present in Africans) recognizes several HLA-C2 molecules [36]. Unlike iKIRs, which educate and enhance the effector activity of NK cells, signaling through aKIRs induces hyporesponsiveness [28,29]. In fact, aKIR/HLA interactions seem to have detrimental effects on immunosurveillance against hematopoietic malignancies, with patients showing increased susceptibility to cancer and/or worse clinical outcomes [37,38,39]. However, these results are contradictory with others that report survival benefits in patients with AML who receive donor grafts with KIR B-haplotypes, rich in aKIRs [40].
Although precise mechanisms have not yet been elucidated, education via aKIRs shares features with the hyporesponsiveness induced by chronic stimulation through the activating receptor NKG2D when interacting with its ligands, the major histocompatibility complex class I chain-related proteins A and B (MICA/B), and the UL16-binding protein (ULBP)-1. Sustained interaction of NKG2D receptor with its ligands expressed on tumor cells has been reported to decrease the association of NKG2D with DAP-10 and KARAP/DAP-12 adaptor proteins by uncoupling NKG2D receptor signaling from intracellular calcium mobilization and decreasing NK cell-mediated cytolysis [41]. Nonetheless, efficient tumor cell killing requires additional activating signaling, such as that of DNAM-1 (CD226) interacting Nectin-2 (CD112) and poliovirus receptor (PVR or CD155) ligands. Although the clinical implications of these interactions have been poorly studied, alterations in DNAM-1 expression lead to higher relapse rates in AML [42] and higher susceptibility to solid and hematologic cancers [43,44,45,46].
NK cells exhibit a multitude of receptors capable of transmitting activating and inhibitory signals when interacting with their specific ligands [47], and therefore, a holistic approach is necessary for a better understanding of their antitumor response. In this study, however, we analyzed the expression of ligands for NK cell receptors that, according to our own experience, play a key role in NK cell education and immune surveillance of different types of cancer [48,49,50,51]. For that reason, the expression of HLA-I and HLA-C, main ligands of iKIRs and aKIRs, and that of CD112/CD155 and ULBP-1/MICA/B, ligands of CD226 and NKG2D, respectively, were prospectively analyzed on B-ALL, T-ALL, and AML pediatric leukemic cells to ascertain their role in the outcome of these patients.

2. Results

2.1. Characteristics and Outcome of Patients According to the Type of Acute Leukemia

Table 1 summarizes the biological and clinical characteristics as well as treatment and outcome of patients according to the type of acute leukemia. Mean follow-up was 40.0 ± 31.3 months. Treatment-related mortality and cumulative incidence of relapse were 8.6% and 2.8% for B-ALL, 0.0% and 12.5% for T-ALL, and 6.25% and 31.2% for AML. Five-year event-free survival (5y-EFS) and overall survival (5y-OS) rates were 88.6% and 90.0% for B-ALL, 87.5% and 100% for T-ALL, and 68.8% and 68.8% for AML, respectively. According to the risk status at enrollment, treatment-related mortality and cumulative incidence of relapse were 7.7% and 0.0% for standard-risk, 2.5% and 2.5% for intermediate-risk, and 11.8% and 20.6% for high-risk patients; 5y-EFS and 5y-OS were 92.3% and 92.3% for standard-risk, 95.0% and 97.5% for intermediate-risk, 67.6% and 73.5% for high-risk patients (Figure 1A). Detailed results for each type of leukemia according to patient risk can be seen in Figure 1B.

2.2. Patient Outcome According to the Expression of Ligands for NK Cell Receptors on Leukemic Blast Cells

First, we evaluated the expression of ligands for activating NK cell receptors measured as mean fluorescence intensity (MFI) on normal and leukemic blast cells (Figure 2A). Compared with their normal residual counterparts (normal lymphocytes for ALL and normal granulocytes for AML), leukemic blast cells showed similar levels of MICA/B expression in B-ALL (123.0 ± 13.1 vs. 135.9 ± 11.1 MFI, n.s.), and T-ALL (119.5 ± 34.0 vs. 172.6 ± 37.6 MFI, n.s.), but lower in AML (394.1 ± 49.3 vs. 316.9 ± 120.1 MFI, p = 0.013); higher levels of ULBP-1 in B-ALL (73.8 ± 10.0 vs. 263.1 ± 42.5 MFI, p < 0.001), T-ALL (126.7 ± 38.5 vs. 251.5 ± 46.8 MFI, n.s.), and AML (636.2 ± 119.6 vs. 719.2 ± 300.0 MFI, p = 0.046); higher levels of CD112 in B-ALL (340.8 ± 48.8 vs. 1868 ± 140.6 MFI, p < 0.001), T-ALL (350.4 ± 84.4 vs. 914.3 ± 245.0 MFI, n.s.), and AML (13,154 ± 345 vs. 3422 ± 476.5 MFI, p < 0.001); and higher levels of CD155 in B-ALL (99.1 ± 6.0 vs. 417.8 ± 128.0 MFI, p < 0.001), and T-ALL (170.1 ± 44.2 vs. 415.0 ± 80.3 MFI, p = 0.026), but similar in AML (1029 ± 137.2 vs. 946.2 ± 114.5 MFI, n.s.).
When comparing the expression of ligands for inhibitory NK cell receptors with that of their normal counterpart cells (Figure 2B), leukemic blast cells showed higher expression of total HLA-I in B-ALL (38,277 ± 3708 vs. 45,314 ± 4975 MFI, n.s.) and AML (37,187 ± 9152 vs. 51,888 ± 9623 MFI, n.s.) but lower in T-ALL (37,122 ± 7468 vs. 17,559 ± 4387 MFI, p = 0.046); and lower expression of HLA-C in B-ALL (14,320 ± 1489 vs. 10,474 ± 1212 MFI, p = 0.028) and T-ALL (12,412 ± 2509 vs. 6861 ± 1832 MFI, p = 0.019) but similar levels in AML (7828 ± 1453 vs. 8877 ± 1523 MFI, n.s.). However, when studying patients individually, we detected lower expression (loss of HLA) on leukemic blasts compared to their normal counterpart cells for HLA-I in 42.4%, 93.3%, and 25.0% (p = 0.0002) and HLA-C in 75.8%, 86.7%, and 62.5% (p = 0.292) of B-ALL, T-ALL, and AML patients, respectively (Figure 3A).
Next, we explored the impact of NK cell receptor ligand expression on outcome of acute leukemia patients. Reduced 5y-EFS rate was observed for patients showing loss of HLA-I (78.3% vs. 90.2%, p = 0.115) and HLA-C (80.8% vs. 95.8%, p = 0.091) on leukemic blast cells compared to patients with maintained or increased expression of these ligands, although these results were not statistically significant (Figure 3B). Regarding ligands for activating NK cell receptors, reduced 5y-EFS rate was observed for patients showing high vs. low expression (cutoff = 140.1) of ULBP-1 (77.6% vs. 94.9%, p = 0.03). However, there were no significant differences in the 5y-EFS rate of patients showing high or low expression of MICA/B (cutoff = 108.5; 79.7% vs. 92.1%, p = 0.126), CD112 (cutoff = 1640; 84.5% vs. 84.6%, p = 0.903), or CD155 (cutoff = 326.1; 83.6% vs. 86.1%, p = 0.652) (Figure 3C).

2.3. Role of KIR/HLA-Ligand Interactions in the Outcome of Acute Leukemia

To investigate the role of NK cell receptor interactions with their ligands expressed on leukemic blast cells, we first explored the role of HLA-I allotypes in the susceptibility to childhood acute leukemia by analyzing their frequency in control and patient groups (Table S1). Only HLA-B*35 was significantly more frequent in childhood acute leukemia than in controls (34.0% vs. 12.0%, p = 0.0005, Pc = 0.009). Since HLA-B*35 is not a Bw4 allele and its interaction with any NK cell receptor has not been reported, we will evaluate its potential role in patient outcome in the following sections of this manuscript. HLA-C*04 also showed higher frequency in patients than in controls (33.3% vs. 20.5%, p = 0.037, Pc = 0.185) but its statistical significance was lost after correction.
Next, we evaluated the frequency of inhibitory and activating KIR receptors, HLA-I ligands, and specific KIR/HLA-I ligand interactions in controls and in total patients, as well as in B-ALL, T-ALL, and AML patients independently (Table 2). No differences in the frequency of KIR genes, HLA-I ligands (HLA-C1, -C2, -A*03/A*11, -Bw4, Bw4-80T, Bw4-80I, or KIR2S4-ligands) or KIR/HLA-I ligand interactions were found between controls and patients with any type of acute leukemia, except for a higher frequency of KIR2DS4-ligands in total patients than in controls (71.0% vs. 53.0%, p = 0.009, Pc = 0.045).
Finally, we evaluated the impact of KIR/HLA-I ligand interactions in the outcome of acute leukemia patients. Specifically, only the presence of the KIR2DL1/C2-ligand interaction was associated with lower 5y-EFS rates (80.0% vs. 96.5%, p = 0.032) in total patients (Figure 4A left, B), as well as in B-ALL (84.8% vs. 95.8%, n.s.), T-ALL (80.0% vs. 100%, n.s.) and AML (64.3% vs. 100%, n.s.) (Figure 4B). To clarify the role of this interaction, we subsequently analyzed the interaction of KIR2DL1 with each of the HLA-C allotypes included in the C2-epitope group (HLA-C*02, *04, *05, *06, *15, *17, *18) (Figure 4A right). It was noteworthy that specifically KIR2DL1/HLA-C*04 interaction was associated with the most reduced 5y-EFS rate (70.6% vs. 92.6%, p < 0.002) and 5y-OS rate (73.5% vs. 95.6%, p = 0.001) in all patients (Figure 4C). Furthermore, KIR2DL1/HLA-C*04 interaction was associated with a higher rate of relapse (17.6% vs. 4.4%, p = 0.035) compared to patients without this interaction. Patients with the KIR2DL1/HLA-C*04 interaction showed the most reduced 5y-EFS rates also in B-ALL (77.3% vs. 93.8%, p = 0.036), T-ALL (75.0% vs. 91.7%, n.s.) and AML (50.0% vs. 87.5%, p = 0.05) patients. Apparently, the KIR2DL1/HLA-C*05 interaction was associated with a better 5y-EFS rate (100% vs. 82.8%), neutralizing the negative impact of KIR2DL1/HLA-C*04 interaction. Thus, only in the absence of HLA-C*05, KIR2DL1/HLA-C*04 interaction showed the most reduced 5y-EFS rate (60.0%, p = 0.0001) compared to that of patients with the KIR2DL1/HLA-C*04 interaction in the presence of HLA-C*05 (100%) or to those without the KIR2DL1/HLA-C*04 interaction whether (96.0%) or not (90.7%) patients were HLA-C*05 (Figure 4D).
In Section 2.3 of this manuscript, we described that HLA-B*35 was significantly associated with acute leukemia. However, as shown in Figure S1, it was mainly due to its association with HLA-C*04, which may also result in HLA-B*35 patients having a poorer outcome due to the KIR2DL1/HLA-C*04 interaction. Furthermore, HLA-B*35 did not add any independent prognostic value to the EFS of patients (HR = 1.148, p = 0.837) when analyzed together with risk status at enrollment (HR = 2.751, p = 0.016) and KIR2DL1/HLA-C*04 interaction (HR = 4.239, p = 0.039, Harrell C-statistic = 0.804 ± 0.052).
Trying to understand why KIR2DL1/HLA-C*04 interaction could be associated with a much lower EFS and OS, the expression of total HLA-I and HLA-C was analyzed on leukemic blast cells of patients carrying the 6 different allotypes with the C2-epitope. Remarkably, only patients with the HLA-C*04 allotype, compared to patients with other C2-epitope allotypes, showed a significant reduction in HLA-C expression on leukemic cells (7391 ± 1046 vs. 10,765 ± 1240 MFI, p = 0.042), but not of HLA-I (13,163 ± 1611 vs. 13,733 ± 1492 MFI, n.s.). The other HLA-C allotypes with the C2-epitope showed maintained or increased expression of HLA-C. Similar results were observed when patients with B-ALL were analyzed independently, with patients who carried the HLA-C*04 allotype showing a significant reduction in HLA-C expression (6903 ± 1412 vs. 12,026 ± 1581 MFI, p = 0.019) compared to patients with other C2-epitope allotypes (Figure 5).
Finally, since HLA C2-epitope is a ligand for KIR2DS1 as well, the role of this activating receptor in the outcome of acute leukemia pediatric patients was evaluated (Figure S2). The number of NK cells expressing KIR2DL1 (18.35% ± 1.48 vs. 18.34% ± 1.76) or KIR2DS1 (11.4% ± 1.07 vs. 11.4% ± 1.08) in the peripheral blood of patients with or without the KIR2DL1/HLA-C*04 interaction showed no differences (Figure S2A). Furthermore, the presence of KIR2DS1 did not alter the differential EFS curves in patients with or without the KIR2DL1/HLA-C*04 interaction (Figure S2B). KIR2DS1 did not add an independent prognostic value to the EFS of patients (HR = 1.373, p = 0.599).

2.4. KIR2DL1/C*04 Interaction Is an Independent Prognostic Biomarker That Complements Risk Status at Enrollment

Cox regression multivariate analysis of total acute leukemia patients showed that KIR2DL1/HLA-C*04 interaction was an independent prognostic factor for EFS (HR = 4.795, p = 0.005, Harrell C-statistic = 0.798 ± 0.051) and OS (HR = 6.731, p = 0.005, Harrell C-statistic = 0.837 ± 0.047), as was the risk status at enrollment (HR = 2.209, p = 0.013 for EFS and HR = 2.182, p = 0.033 for OS) when analyzed together with sex, age, the type of leukemia and the low/high expression of ULBP-1 on leukemic blast cells (Figure 6A).
Indeed, the presence of the KIR2DL1/C*04 interaction complemented the risk status at patient enrollment whether they were new diagnoses or relapses. Although similar suitable 5y-EFS rates were observed for standard-/intermediate-risk patients according to the absence/presence of the KIR2DL1/C*04 interaction (95.6% vs. 90.5%), a much lower 5y-EFS rate was observed in high-risk patients in the presence of the KIR2DL1/C*04 interaction (38.5% vs. 85.7%, p < 0.002) (Figure 6B). The negative prognostic value of the absence/presence of the KIR2DL1/C*04 interaction was maintained both in patients enrolled at diagnosis (5y-OS rate of 96.9% vs. 82.8%, p < 0.015) and in patients enrolled at relapse (5y-OS rate of 66.7% vs. 20.0%, n.s.), although in relapsed patients results were not statistically significant due to their reduced number (n = 8) (Figure 6C).

2.5. Models to Predict NK Cell Alloreactivity in Allogeneic HSCT in Our Series

Nineteen allogeneic HSCTs in 18 high-risk patients (7 AML, 8 B-ALL, and 3 T-ALL) were performed with 6 haploidentical, 5 related (RD), and 8 unrelated (UD) donors (Table S2). Seven out of 19 HSCT patients relapsed (36.8%), and 6 died (31.5%). According to donor type, we found 66.7%, 40.0%, and 12.5% cumulative incidence of relapse and 50.0%, 20.0%, and 25% incidence of death for haploidentical, RD, and UD donors, respectively (Table S2, up).
KIR2DL1/HLA-C*04 interaction was present in 4 out of 6 (66.7%) patients who relapsed and in 66.7% of their donors, as well as in 6 out of 6 (100%) patients who died and in 83.3% of their donors. It is important to bear in mind that the frequency of KIR2DL1/HLA-C*04 interaction in healthy controls from our series was 19.3%. Nonetheless, KIR2DL1/HLA-C*04 interaction was also present in 7 out of 12 (58.3%) patients without relapse and in 50.0% of their donors, as well as in 6 out of 13 (46.15%) patients who survived and in 38.4% of their donors.
According to models used to predict NK cell alloreactivity [53], the receptor-ligand model showed increasing cumulative incidence of relapse and death in donor-recipient pairs with missing ligands for KIR2DL1 (n = 1, 0% and 0%), KIR2DS1 (n = 4, 25% and 0%), KIR3DL2 (n = 13, 31% and 23%), KIR3DL1 (n = 13, 54% and 38%) and KIR2DL2/L3 (n = 4, 50% and 50%). For the ligand-incompatibility model increasing cumulative incidence of relapse and death were found in donor-recipient pairs with licensed KIR2DS1 (n = 4, 25% and 0%) and KIR3DL1 (n = 6, 50%, and 33%), with the worst results for those without licensed KIRs for missing ligands (n = 9, 44% and 44%). Although the small number of cases did not allow us to draw strong conclusions, donor-recipient pairs with functional KIR2DS1 (donor no C2-homozygous and recipient bearing its C2 ligands [29]) showed the best results (Table S2, down).

3. Discussion

Maintenance therapy with 6-mercaptopurine and methotrexate is critical to ensure the cure of most cases of childhood acute lymphoblastic leukemia. The antileukemic effect of this therapy is primarily mediated by DNA incorporation of thioguanine nucleotides that favor nucleotide mismatching, which causes apoptosis after futile attempts of mismatch repair [54]. It is also suspected that immunological effectors may play a role in the eradication of tumor cells during this phase. Although these mechanisms are largely unknown, the high diversity of molecules guiding the interactions of NK cells with their tumor targets make them suitable candidates to play a role in tumor immune surveillance during this period, and it could help explain outcome variability among patients. In fact, NK cells are the first lymphoid cell to recover after therapy [55,56] and could be involved in the elimination of cancer cells. In support of this hypothesis, numerous studies have shown that HSCT with the appropriate haploidentical donor may improve clinical outcomes [8,9,57,58,59]. The main effectors in haploidentical HSCT are NK cells, which trigger their antitumor activity in case of missing ligand in the leukemic cells [8,9]. Our results show how the expression of ligands for the receptors that regulate NK cell function on leukemic cells is associated with the clinical outcome and survival of patients with ALL and AML. Interactions of these receptors, specifically KIR2DL1 with HLA-C*04 allotype (bearing the C2-epitope), have an independent predictive value and might complement the current risk stratification used in the clinical management of pediatric patients, particularly in those with high-risk features. This could contribute to personalizing not only the type of induction and consolidation therapy but also the intensity/length of maintenance therapy. Maintenance therapy, although less intensive, does have risks, as patients can develop life-threatening infections and late effects from prolonged cytotoxicity [60]. Therefore, children at lower immunological risk of events or death could benefit from reduced or, on the contrary, from increased maintenance treatment, whereas high immunological risk children could benefit from alternative therapies.
Recent evidence demonstrates that thiopurine-induced drug resistance mutations facilitate pediatric ALL relapses in a subset of patients, suggesting that decreasing exposure to these drugs by shortening the maintenance period might be beneficial because it reduces the chance of developing chemoresistant clones [61]. However, this reduction should be based on appropriate biomarkers because attempts to reduce the duration of maintenance therapy have generally led to an increase in relapses. Results from our series of pediatric acute leukemia show a clear association between relapse and the presence of the KIR2DL1/HLA-C*04 interaction, present in 17.6% of patients with relapse and only in 4.4% of patients without relapse, which highlights the possible participation of immune mechanisms in this process. Furthermore, our data reveal that leukemic cells of patients with the KIR2DL1/HLA-C*04 interaction have a selective loss in the expression of HLA-C but not of total HLA-I. Although the DT9 antibody used to assess the expression of HLA-C in our series may recognize both HLA-C and HLA-E; apparently, HLA-C is the antigen primarily detected by the antibody because HLA-E is poorly expressed in peripheral blood lymphocytes [62]. Unfortunately, our results do not allow us to determine whether the loss of HLA-C expression is causative of relapse and/or death of patients or if the surveillance pressure exerted by NK cells through the KIR2DL1/HLA-C*04 interaction could contribute to selecting more aggressive leukemic clones that specifically reduce the expression of HLA-C (and increase ligands for NK receptors such as ULBP-1) to escape the immune response. Our results do not allow us either to establish whether the effectors driving this phenomenon are NK cells or cytolytic T cells through the recognition of tumor peptides presented in HLA-C*04. This fact, however, seems less likely given that the negative effect of the KIR2DL1/HLA-C*04 interaction was observed in different lineages of acute leukemia, lymphoblastic (B and T), and myeloblastic. Yet, in turn, the protection that the KIR2DL1/C*05 interaction seems to exert against the detrimental effect of the KIR2DL1/HLA-C*04 interaction suggests the involvement of specific immunological mechanisms. In any case, these results are in line with those describing that peptide selectivity of KIR/Ligand interaction would allow NK cells to respond, not only to changes in the surface expression of HLA-C but also to the more subtle changes in the HLA-C peptidome [19].
Although the molecular mechanisms involved in the negative effect of the KIR2DL1/HLA-C*04 interaction need to be elucidated, there are important precedents showing that: (1) KIR2DL1/C2-epitope interaction is associated with increased susceptibility to childhood ALL and with an elevated risk of late relapse [63]; (2) frequency of HLA-Cw4 is elevated in patients with ALL (relative risk = 2.01, p < 0.0003), AML (relative risk = 2.06, p < 0.0002), and CML (relative risk = 2.14, p < 0.0008) [64]; (3) HLA-Cw4 is associated with ALL [65,66]; (4) C2-epitope is associated with reduced EFS in patients with B-CLL, particularly in KIR2DS1 negative patients, whose only receptor for the C2-epitope is KIR2DL1 [67]; and (5) presence of C2-epitope is associated with increased risk of relapse after HLA-matched transplantation in patients with acute and chronic myeloid leukemia, irrespective of donor relation [68]. Unfortunately, the aforementioned study did not evaluate the role of the C2-epitope at specific HLA-C allotype levels. Although the small number of patients who received HSCT in our series did not allow us to adequately explore the relationship of donor-derived KIR/HLA interactions with the patient’s bones, according to previous studies [68]. KIR2DL1/HLA-C*04 interaction was associated with poor outcomes, irrespective of the type of the allogenic HSCT (haploidentical, related, or unrelated) performed in our high-risk relapsed patients. Since the KIR2DL1/HLA-C*04 genotype seems to favor the escape of leukemic clones refractory to conventional and NK cell therapies (HSCT), these children should be candidates for alternative treatments such as BiTe, CART, or experimental treatment for AML. On the contrary, and although the associated mechanisms are unknown, children who carry the HLA-C*05 allotype seem to have more favorable outcomes and would be suitable candidates for NK cell therapy in the unlikely event of relapse, since relapse in children with this genotype is rare.
An independent study including most ALL pediatric patients from our series reported that persistent secondhand smoke (SHS) of smoking parents worsens OS and treatment-related mortality in these children [69]. Authors described that the main cause of death associated with SHS was an infection, which poses a new question for our study: could the KIR2DL1/HLA-C*04 interaction be related to defective protection against infections instead of to an altered antitumor response? However, since KIR2DL1/HLA-C*04 interaction was associated with a higher rate of relapse, immunosuppression caused by intensification treatments could be the main reason for infection and death. Nonetheless, to answer this question, future biological studies on a new and larger series of pediatric patients with acute leukemia will be necessary.
Regarding the expression of ligands for NK cell activating receptors, our results for ULBP-1, a ligand of NKG2D, are in disagreement with those reported for AML, in which a higher expression of ULBP-1 was associated with reduced relapse and improved 2-year overall survival [70]. This supports, in contrast, that sustained interaction of NKG2D receptor with its ligands expressed on tumor cells decreases NK cell-mediated cytolysis by uncoupling NKG2D receptor signaling from intracellular calcium mobilization [41]. However, in our series, these data had no independent prognostic value when compared with the risk at enrollment and the presence of the KIR2DL1/HLA-C*04 interaction. MICA/B expression shows marginal no significant changes between leukemic and normal cells. However, a higher expression of this NKG2D ligand was associated with reduced EFS, which is in agreement with the reduced NK cell function associated with reduced expression of activating NK receptors in high-risk myelodysplastic syndrome [71]. In contrast, the expression of DNAM-1 ligands (CD112 and CD155) showed the most significant increases in leukemic cells compared to normal cells, but this had no impact on the survival of patients. This suggests that, in pediatric acute leukemia, effective NK cell immune surveillance might depend on NKG2D more than on DNAM-1.
Collectively, our results show that the expression of ligands for the receptors that regulate the function of NK cells on leukemic cells is associated with the clinical outcome of patients with ALL and AML. Interactions of these receptors, specifically KIR2DL1 with HLA-C*04 allotype (bearing the C2-epitope), have an independent predictive value and might complement the current risk stratification used in the clinical management of pediatric patients. This could contribute to personalizing the type and intensity of therapy. Since the KIR2DL1/HLA-C*04 genotype seems to favor the escape of leukemic clones refractory to conventional and HSCT NK cell-based therapies, these children should be candidates for alternative treatments such as BiTe, CART, or experimental AML treatment.

4. Materials and Methods

4.1. Patients and Samples

This prospective observational study included 83 sex and aged-matched healthy controls and 102 consecutive (from 2012 to 2020) pediatric patients with B acute lymphoblastic leukemia (B-ALL, n = 70), T acute lymphoblastic leukemia (T-ALL, n = 16), and acute myeloid leukemia (AML, n = 16) from the Clinic University Hospital Virgen de la Arrixaca (Murcia, Spain) (Table 1). The institutional review board (IRB-00005712) approved the study. Written informed consent was obtained from all patients and controls (or their parents) in accordance with the Declaration of Helsinki.
EDTA anti-coagulated peripheral blood (PB) samples for HLA-I and KIR genotyping were obtained from healthy children randomly selected from unrelated volunteers, without chronic diseases, and with a negative family history of hereditary diseases, autoimmune diseases, or malignancies.
EDTA anti-coagulated BM samples were obtained from all patients, at diagnosis or at relapse, to perform cytomorphology, immunophenotype, cytogenetics (karyotype and fluorescence in situ hybridization, FISH), and molecular studies. KIR ligand expression was also evaluated on leukemic blast cells from these BM samples. Diagnostic criteria for the type and subtype of acute leukemia were based on the WHO classification of tumors of hematopoietic and lymphoid tissues [72]. Patient risk was estimated according to age, peripheral blood leukocyte count, immunophenotype, infiltration of central nervous system or testes, cytogenetic abnormalities, sensitivity to prednisone, and bone marrow response at day +15, +33, and +78. Following criteria of the SEHOP_PETHEMA_2013 protocol (https://www.sehh.es/images/stories/recursos/2014/documentos/guias/LAL_SEHOP_PETHEMA_2013.pdf (accessed on 15 January 2021)), ALL patients were classified into standard, intermediate or high risk. In AML patients, risk was classified as standard or high following the criteria of the NOPHO-DBH AML-2012 Protocol (https://www.skion.nl/workspace/uploads/NOPHO-DBH-AML-2012_Protocol-v2-1_17-01-2013.pdf (accessed on 15 January 2021)).
Depending on patient risk, ALL was treated according to the SEHOP-PETHEMA 2013 protocol or the previous PETHEMA 2005 protocol. In these protocols, all patients with T-ALL received L-asparagine depletion during maintenance and intensification/reinduction phases (https://www.fundacionpethema.es/2016/12/12/novedades-lal-sehop-pethema/ (accessed on 15 January 2021)). AML was treated according to the SHOP-LMA 2007 protocol or the most recent NOPHO-DBH AML 2012 protocol. For hematopoietic stem cell transplantation (HSCT), patients received a conditioning regimen followed by the infusion of donor cells. Patients remained hospitalized until hematopoietic and clinical recovery (details on Table 1).

4.2. KIR and HLA Genotyping

KIR and HLA-A, -B, and -C genotyping were performed in DNA samples extracted with QIAmpDNABlood Mini kit (QIAgen, GmbH, Hilden, Germany) using sequence-specific oligonucleotide PCR (PCR-SSO) and Luminex® technology with Lifecodes KIR-SSO (cat. 545110R) and LifecodesHLA-SSO HLA-A (cat. 628913), HLA-B (cat. 628917) and HLA-C (cat. 628921) typing kits (Immucor Transplant Diagnostic, Inc. Stamford, CT, USA), as previously described [48,49]. HLA-A and HLA-B genotyping allowed us to identify alleles bearing the Bw4 motif according to the amino-acid sequences at positions 77–83 in the α1 domain of the HLA class I heavy chain. Bw4 alleles with threonine at amino acid 80 (80T) and higher affinity for KIR3DL1 (HLA-B*05, B*13, B*44) were distinguished from those with isoleucine 80 (I80) and lower affinity (HLA-A*23, A*24, A*25, A*32 and HLA-B*17, B*27, B*37, B*38, B*47, B*49, B*51, B*52, B*53, B*57, B*58, B*59, B*63, and B*77). HLA-C genotyping allowed distinction between HLA-C alleles with asparagine 80 (C1-epitope: HLA-C*01, 03, 07, 08, 12, 14, 16:01) and lysine 80 (C2-epitope: HLA-C*02, *04, *05, *06, *15, *16:02, *17, *18) [24]. Nonetheless, the KIR ligand calculator at https://www.ebi.ac.uk/ipd/kir/ligand.html (accessed on 15 January 2021) was used to ascertain Bw4, C1, and C2 epitopes.
KIR genotyping identified iKIRs (KIR2DL1, KIR2DL2, KIR2DL3, KRIR2DL5, KIR3DL1, KIR3DL2, and KIR3DL3) and aKIRs (KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, and KIR3DS1), as well as KIR2DL4, which shows both inhibitory and activating potential. The method used could not distinguish between KIR2DL5A (telomeric) and KIR2DL5B (centromeric) forms. Different allotypes of KIR2DS4 were detected, including the expressed allotype KIR2DS4 full-length exon-5 (KIR2DS4f, KIR2DS4*001-002 alleles) and the non-expressed KIR2DS4 deleted exon-5 (KIR2DS4d, KIR2DS4*003, *004, *006, *007, and *008 alleles).

4.3. Immunophenotyping of Normal Cells and Leukemic Blasts

The expression of ligands for activating (MICA/B, ULBP-1, CD112, and CD155) and inhibitory (HLA-I and HLA-C) NK cell receptors were evaluated as mean fluorescence intensity (MFI) on BM normal residual cells and on leukemic blast cells using FACSCanto-II and BD FACSDiVa™ Software (Becton Dickinson, BD, San Diego, CA, USA). Photomultiplier (PMT) voltages were adjusted daily using CS&T beads (BD, Ref. 656047). Fluorescence compensations were adjusted every two months using blood samples stained for simple fluorochromes with conjugated anti-CD4 antibodies for each cytometer detector and then re-adjusted finely during the analysis using negative events as reference for each fluorochrome. Briefly, 100µL of BM samples diluted with PBS-1%BSA to contain 0.5 million total white cells were labeled in 5 tubes, all of them containing CD45-APCCy7 (BD, Ref. 348815) and, depending on the type of leukemia, CD7-PECy7 (BD, Ref. 564019) for T-ALL, CD19-PECy7 (BD, Ref. 341113) for B-ALL or CD34 PE-Cy7 (BD, Ref. 348811) or CD33-PECy7 (BD, Ref. 333946) for AML, so as to detect total lymphocytes, granulocytes and leukemic blast cells following the gating strategy described in Figure 7. Then, we added: (1) for tube-1, isotype-γ2a-FITC and isotype-γ1-PE (BD, Ref. 340394), isotype-γ2a-APC (Biolegend, Ref. 400219), and isotype-γ2b-BV421 (BD, Ref. 562748); (2) for tube-2, HLA-I-FITC (Bio-Rad, AbDSerotec, Kidlington, U.K., Ref. MCA81F); (3) for tube-3, purified anti-HLA-C (clone DT9, kindly provided by Dr. Simon Brackenridge from Oxford University, U.K.), which was revealed with a polyclonal anti-IgG (minimal x-reactivity)-BV421 (Biolegend, Ref. 405317) and then CD112 (Nectin-2)-PE (BD, Ref. 337410); (4) for tube-4, CD155 (PVR)-PE (BD, Ref. 337610); and (5) for tube-5, ULBP-1-PE (R&D Systems, Minneapolis, MN, USA, Ref. FAB1380P) and MICA/B-APC (BioLegend, San Diego, CA, USA, Ref. 320908).
MFI of lymphocytes, granulocytes, and leukemic blast cells in the FITC, PE, APC, and V450 (BV421) channels were estimated in the isotype tube, and these MFIs (background) were subtracted from the MFI values estimated for CD112, CD155, and ULBP-1 in the PE-channel, MICA/B in the APC-channel, HLA-I in the FITC-channel and HLA-C in the V450 channel for leukocyte subsets, respectively.

4.4. Statistical Analysis

All data were collected in a database (Excel2003; Microsoft Corporation, Seattle, WA, USA) and analyzed with IBM SPSS Statistics v15.0 (SPSS Inc., Chicago, IL, USA). Student t-test and ANOVA with post hoc tests were used to analyze continuous variables. Data were expressed as mean ± SEM. Kaplan–Meier and log-rank tests were used to analyze patient event-free survival (EFS) and overall survival (OS). Time to event or death was estimated in months from the date of enrollment. Multivariate analyses of prognostic factors for EFS and OS were performed using the Cox proportional hazards model (stepwise regression). Hazard ratio (HR) and 95% confidence interval were estimated. Harrell’s C-statistic was obtained using STATA-14 (Somersd package). p < 0.05 was considered statistically significant. The Bonferroni correction (Pc) was applied when needed.

5. Conclusions

Expression of ligands for activating and inhibitory receptors of NK cells on leukemic cells is associated with the clinical outcome of patients with ALL and AML. Specifically, reduced expression of HLA-C on leukemic cells of patients with the KIR2DL1/HLA-C*04 interaction was associated with a higher rate of relapse and lower patient survival. KIR2DL1/HLA-C*04 interaction was an independent prognostic factor that complemented the risk stratification used in the therapeutic management of pediatric patients.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cancers13102294/s1, Figure S1: Reduced event-free survival (EFS) of acute leukemia patients associated with HLA-B*35 is due to its linkage disequilibrium with HLA-C*04; Figure S2: KIR2DS1 did not alter the event-free survival (EFS) curves of acute leukemia pediatric patients according to the presence of KIR2DL1/HLA-C*04 interaction; Table S1: HLA-I allele frequencies in controls and patients; Table S2: Allogeneic stem cell transplantation performed in our series; File S1: Primary data of patients.

Author Contributions

Conceptualization, A.M. and J.L.F.; methodology, M.V.M.-S., J.A.C., A.M.G., M.B.-C., M.E.L., E.R.-E., J.F.P.-G., A.M.F., L.G., H.M.-B., J.A.G., and M.M.; validation, A.M., M.V.M.-S., and J.L.F.; formal analysis, A.M. and M.V.M.-S.; investigation, A.M., J.L.F., and M.V.M.-S.; resources, A.M., J.L.F., and M.M.; data curation, A.M.; writing—original draft preparation, A.M. and M.V.M.-S.; writing—review and editing, A.M.; visualization, all authors; supervision, A.M. and J.A.C.; project administration, A.M. and J.L.F.; funding acquisition, A.M. and J.L.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Asociación Pablo Ugarte (APU) and M.V.M.-S. was funded by APU.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of Clinic and University Hospital Virgen de la Arrixaca, Murcia, Spain (IRB-00005712).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study or their legal representatives.

Data Availability Statement

The data presented in this study are openly available in File S1.

Acknowledgments

We would like to acknowledge the hematologists involved in the bone marrow transplantation of patients Miguel Blanquer, José María Moraleda and Andrés Sánchez-Salinas, nurses and other health professionals from the pediatric hematology and oncology department, laboratory technicians María Carmen Martínez Solano, María Dolores García Arnao, and María Elena Bernal Moreno, and especially to all patients and families.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lonetti, A.; Pession, A.; Masetti, R. Targeted Therapies for Pediatric AML: Gaps and Perspective. Front. Pediatr. 2019, 7, 463. [Google Scholar] [CrossRef]
  2. Teachey, D.T.; Hunger, S.P.; Loh, M.L. Optimizing therapy in the modern age: Differences in length of maintenance therapy in acute lymphoblastic leukemia. Blood 2021, 137, 168–177. [Google Scholar] [CrossRef] [PubMed]
  3. Bhojwani, D.; Pui, C.-H. Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 2013, 14, e205-17. [Google Scholar] [CrossRef]
  4. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  5. von Stackelberg, A.; Locatelli, F.; Zugmaier, G.; Handgretinger, R.; Trippett, T.M.; Rizzari, C.; Bader, P.; O’Brien, M.M.; Brethon, B.; Bhojwani, D.; et al. Phase I/Phase II Study of Blinatumomab in Pediatric Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2016, 34, 4381–4389. [Google Scholar]
  6. Bhojwani, D.; Sposto, R.; Shah, N.N.; Rodriguez, V.; Yuan, C.; Stetler-Stevenson, M.; O’Brien, M.M.; McNeer, J.L.; Quereshi, A.; Cabannes, A.; et al. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Leukemia 2019, 33, 884–892. [Google Scholar] [CrossRef]
  7. Fuster, J.L.; Molinos-Quintana, A.; Fuentes, C.; Fernández, J.M.; Velasco, P.; Pascual, T.; Rives, S.; Dapena, J.L.; Sisinni, L.; López-Godino, O.; et al. Blinatumomab and inotuzumab for B cell precursor acute lymphoblastic leukaemia in children: A retrospective study from the Leukemia Working Group of the Spanish Society of Pediatric Hematology and Oncology (SEHOP). Br. J. Haematol. 2020, 190, 764–771. [Google Scholar] [PubMed]
  8. Davies, S.M.; Iannone, R.; Alonzo, T.A.; Wang, Y.-C.; Gerbing, R.; Soni, S.; Kolb, E.A.; Meshinchi, S.; Orchard, P.J.; Burns, L.J.; et al. A Phase 2 Trial of KIR-Mismatched Unrelated Donor Transplantation Using in Vivo T Cell Depletion with Antithymocyte Globulin in Acute Myelogenous Leukemia: Children’s Oncology Group AAML05P1 Study. Biol. Blood Marrow Transplant. 2020, 26, 712–717. [Google Scholar] [PubMed]
  9. Mamcarz, E.; Madden, R.; Qudeimat, A.; Srinivasan, A.; Talleur, A.; Sharma, A.; Suliman, A.; Maron, G.; Sunkara, A.; Kang, G.; et al. Improved survival rate in T-cell depleted haploidentical hematopoietic cell transplantation over the last 15 years at a single institution. Bone Marrow Transplant. 2020, 55, 929–938. [Google Scholar]
  10. Rubnitz, J.E.; Inaba, H.; Kang, G.; Gan, K.; Hartford, C.; Triplett, B.M.; Dallas, M.; Shook, D.; Gruber, T.; Pui, C.-H.; et al. Natural killer cell therapy in children with relapsed leukemia. Pediatr. Blood Cancer 2015, 62, 1468–1472. [Google Scholar] [CrossRef] [Green Version]
  11. Fernández, A.; Navarro-Zapata, A.; Escudero, A.; Matamala, N.; Ruz-Caracuel, B.; Mirones, I.; Pernas, A.; Cobo, M.; Casado, G.; Lanzarot, D.; et al. Optimizing the Procedure to Manufacture Clinical-Grade NK Cells for Adoptive Immunotherapy. Cancers 2021, 13, 577. [Google Scholar] [CrossRef] [PubMed]
  12. Yanir, A.D.; Martinez, C.A.; Sasa, G.; Leung, K.; Gottschalk, S.; Omer, B.; Ahmed, N.; Hegde, M.; Eunji, J.; Liu, H.; et al. Current Allogeneic Hematopoietic Stem Cell Transplantation for Pediatric Acute Lymphocytic Leukemia: Success, Failure and Future Perspectives-A Single-Center Experience, 2008 to 2016. Biol. Blood Marrow Transplant. 2018, 24, 1424–1431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Moretta, L.; Bottino, C.; Pende, D.; Castriconi, R.; Mingari, M.C.; Moretta, A. Surface NK receptors and their ligands on tumor cells. Semin. Immunol. 2006, 18, 151–158. [Google Scholar] [PubMed]
  14. Kärre, K. Natural killer cell recognition of missing self. Nat. Immunol. 2008, 9, 477–480. [Google Scholar] [CrossRef] [PubMed]
  15. Parham, P. MHC class I molecules and KIRS in human history, health and survival. Nat. Rev. Immunol. 2005, 5, 201–214. [Google Scholar] [CrossRef] [PubMed]
  16. Middleton, D.; Vilchez, J.R.; Cabrera, T.; Meenagh, A.; Williams, F.; Halfpenny, I.; Maleno, I.; Ruiz-Cabello, F.; Lopez-Nevot, M.A.; Garrido, F. Analysis of KIR gene frequencies in HLA class I characterised bladder, colorectal and laryngeal tumours. Tissue Antigens 2007, 69, 220–226. [Google Scholar] [CrossRef]
  17. Ruggeri, L.; Mancusi, A.; Perruccio, K.; Burchielli, E.; Martelli, M.F.; Velardi, A. Natural killer cell alloreactivity for leukemia therapy. J. Immunother. 2005, 28, 175–182. [Google Scholar]
  18. Pende, D.; Falco, M.; Vitale, M.; Cantoni, C.; Vitale, C.; Munari, E.; Bertaina, A.; Moretta, F.; Del Zotto, G.; Pietra, G.; et al. Killer Ig-Like Receptors (KIRs): Their Role in NK Cell Modulation and Developments Leading to Their Clinical Exploitation. Front. Immunol. 2019, 10, 1179. [Google Scholar] [CrossRef] [Green Version]
  19. Hilton, H.G.; Parham, P. Missing or altered self: Human NK cell receptors that recognize HLA-C. Immunogenetics 2017, 69, 567–579. [Google Scholar] [CrossRef]
  20. Enqvist, M.; Ask, E.H.; Forslund, E.; Carlsten, M.; Abrahamsen, G.; Béziat, V.; Andersson, S.; Schaffer, M.; Spurkland, A.; Bryceson, Y.; et al. Coordinated Expression of DNAM-1 and LFA-1 in Educated NK Cells. J. Immunol. 2015, 194, 4518–4527. [Google Scholar] [CrossRef]
  21. Anfossi, N.; André, P.; Guia, S.; Falk, C.S.; Roetynck, S.; Stewart, C.A.; Breso, V.; Frassati, C.; Reviron, D.; Middleton, D.; et al. Human NK Cell Education by Inhibitory Receptors for MHC Class I. Immunity 2006, 25, 331–342. [Google Scholar] [CrossRef]
  22. Béziat, V.; Descours, B.; Parizot, C.; Debré, P.; Vieillard, V. NK cell terminal differentiation: Correlated stepwise decrease of NKG2A and acquisition of KIRs. PLoS ONE 2010, 5, e11966. [Google Scholar] [CrossRef]
  23. Kim, S.; Poursine-Laurent, J.; Truscott, S.M.; Lybarger, L.; Song, Y.J.; Yang, L.; French, A.R.; Sunwoo, J.B.; Lemieux, S.; Hansen, T.H.; et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 2005, 436, 709–713. [Google Scholar] [CrossRef]
  24. Mandelboim, O.; Reyburn, H.T.; Valés-Gómez, M.; Pazmany, L.; Colonna, M.; Borsellino, G.; Strominger, J.L. Protection from lysis by natural killer cells of group 1 and 2 specificity is mediated by residue 80 in human histocompatibility leukocyte antigen C alleles and also occurs with empty major histocompatibility complex molecules. J. Exp. Med. 1996, 184, 913–922. [Google Scholar] [CrossRef] [Green Version]
  25. Foley, B.A.; De Santis, D.; Van Beelen, E.; Lathbury, L.J.; Christiansen, F.T.; Witt, C.S. The reactivity of Bw4+ HLA-B and HLA-A alleles with kir3dll: Implications for patient and donor suitability for haploidentical stem cell transplantations. Blood 2008, 112, 435–443. [Google Scholar] [CrossRef] [PubMed]
  26. Hansasuta, P.; Dong, T.; Thananchai, H.; Weekes, M.; Willberg, C.; Aldemir, H.; Rowland-Jones, S.; Braud, V.M. Recognition of HLA-A3 and HLA-A11 by KIR3DL2 is peptide-specific. Eur. J. Immunol. 2004, 34, 1673–1679. [Google Scholar] [CrossRef]
  27. Horowitz, A.; Djaoud, Z.; Nemat-Gorgani, N.; Blokhuis, J.; Hilton, H.G.; Béziat, V.; Malmberg, K.-J.; Norman, P.J.; Guethlein, L.A.; Parham, P. Class I HLA haplotypes form two schools that educate NK cells in different ways. Sci. Immunol. 2016, 1, eaag1672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Chewning, J.H.; Gudme, C.N.; Hsu, K.C.; Selvakumar, A.; Dupont, B. KIR2DS1-Positive NK Cells Mediate Alloresponse against the C2 HLA-KIR Ligand Group In Vitro. J. Immunol. 2007, 179, 854–868. [Google Scholar] [CrossRef]
  29. Fauriat, C.; Ivarsson, M.A.; Ljunggren, H.; Malmberg, K.; Michae, J. Education of human natural killer cells by activating killer cell immunoglobulin-like receptors. Blood 2010, 115, 1166–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. David, G.; Djaoud, Z.; Willem, C.; Legrand, N.; Rettman, P.; Gagne, K.; Cesbron, A.; Retiere, C. Large Spectrum of HLA-C Recognition by Killer Ig-like Receptor (KIR)2DL2 and KIR2DL3 and Restricted C1 Specificity of KIR2DS2: Dominant Impact of KIR2DL2/KIR2DS2 on KIR2D NK Cell Repertoire Formation. J. Immunol. 2013, 191, 4778–4788. [Google Scholar] [CrossRef] [Green Version]
  31. Liu, J.; Xiao, Z.; Ko, H.L.; Shen, M.; Ren, E.C. Activating killer cell immunoglobulin-like receptor 2DS2 binds to HLA-A*11. Proc. Natl. Acad. Sci. USA 2014, 111, 2662–2667. [Google Scholar] [CrossRef] [Green Version]
  32. Thiruchelvam-Kyle, L.; Hoelsbrekken, S.E.; Saether, P.C.; Bjørnsen, E.G.; Pende, D.; Fossum, S.; Daws, M.R.; Dissen, E. The Activating Human NK Cell Receptor KIR2DS2 Recognizes a β2-Microglobulin-Independent Ligand on Cancer Cells. J. Immunol. 2017, 198, 2556–2567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Graef, T.; Moesta, A.K.; Norman, P.J.; Abi-Rached, L.; Vago, L.; Older Aguilar, A.M.; Gleimer, M.; Hammond, J.A.; Guethlein, L.A.; Bushnell, D.A.; et al. KIR2DS4 is a product of gene conversion with KIR3DL2 that introduced specificity for HLA-A*11 while diminishing avidity for HLA-C. J. Exp. Med. 2009, 206, 2557–2572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Garcia-Beltran, W.F.; Hölzemer, A.; Martrus, G.; Chung, A.W.; Pacheco, Y.; Simoneau, C.R.; Rucevic, M.; Lamothe-Molina, P.A.; Pertel, T.; Kim, T.; et al. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat. Immunol. 2016, 17, 1067–1074. [Google Scholar] [CrossRef] [PubMed]
  35. O’Connor, G.M.; Yamada, E.; Rampersaud, A.; Thomas, R.; Carrington, M.; McVicar, D.W. Analysis of binding of KIR3DS1*014 to HLA Suggests Distinct Evolutionary History of KIR3DS1. J. Immunol. 2011, 187, 2162–2171. [Google Scholar] [CrossRef] [Green Version]
  36. Blokhuis, J.H.; Hilton, H.G.; Guethlein, L.A.; Norman, P.J.; Nemat-Gorgani, N.; Nakimuli, A.; Chazara, O.; Moffett, A.; Parham, P. KIR2DS5 allotypes that recognize the C2 epitope of HLA-C are common among Africans and absent from Europeans. Immun. Inflamm. Dis. 2017, 5, 461–468. [Google Scholar] [CrossRef] [Green Version]
  37. Nowak, J.; Kościńska, K.; Mika-Witkowska, R.; Rogatko-Koroś, M.; Mizia, S.; Jaskuła, E.; Polak, M.; Mordak-Domagała, M.; Lange, J.; Gronkowska, A.; et al. Role of Donor Activating KIR-HLA Ligand-Mediated NK Cell Education Status in Control of Malignancy in Hematopoietic Cell Transplant Recipients. Biol. Blood Marrow Transplant. 2015, 21, 829–839. [Google Scholar] [CrossRef] [Green Version]
  38. Sullivan, E.M.; Jeha, S.; Kang, G.; Cheng, C.; Rooney, B.; Holladay, M.; Bari, R.; Schell, S.; Tuggle, M.; Pui, C.-H.; et al. NK cell genotype and phenotype at diagnosis of acute lymphoblastic leukemia correlate with postinduction residual disease. Clin. Cancer Res. 2014, 20, 5986–5994. [Google Scholar] [CrossRef] [Green Version]
  39. Misra, M.K.; Prakash, S.; Moulik, N.R.; Kumar, A.; Agrawal, S. Genetic associations of killer immunoglobulin like receptors and class I human leukocyte antigens on childhood acute lymphoblastic leukemia among north Indians. Hum. Immunol. 2016, 77, 41–46. [Google Scholar] [CrossRef]
  40. Cooley, S.; Trachtenberg, E.; Bergemann, T.L.; Saeteurn, K.; Klein, J.; Le, C.T.; Marsh, S.G.E.; Guethlein, L.A.; Parham, P.; Miller, J.S.; et al. Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood 2009, 113, 726–732. [Google Scholar] [CrossRef] [Green Version]
  41. Coudert, J.D.; Zimmer, J.; Tomasello, E.; Cebecauer, M.; Colonna, M.; Vivier, E.; Held, W. Altered NKG2D function in NK cells induced by chronic exposure to NKG2D-ligand expressing tumor cells. Blood 2005, 106, 1711–1718. [Google Scholar] [CrossRef] [Green Version]
  42. Khaznadar, Z.; Boissel, N.; Agaugué, S.; Henry, G.; Cheok, M.; Vignon, M.; Geromin, D.; Cayuela, J.-M.; 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, 2580–2590. [Google Scholar] [CrossRef] [Green Version]
  43. Carlsten, M.; Björkström, N.K.; Norell, H.; Bryceson, Y.; Van Hall, T.; Baumann, B.C.; Hanson, M.; Schedvins, K.; Kiessling, R.; Ljunggren, H.G.; et al. DNAX accessory molecule-1 mediated recognition of freshly isolated ovarian carcinoma by resting natural killer cells. Cancer Res. 2007, 67, 1317–1325. [Google Scholar] [CrossRef] [Green Version]
  44. Labani-Motlagh, A.; Israelsson, P.; Ottander, U.; Lundin, E.; Nagaev, I.; Nagaeva, O.; Dehlin, E.; Baranov, V.; Mincheva-Nilsson, L. Differential expression of ligands for NKG2D and DNAM-1 receptors by epithelial ovarian cancer-derived exosomes and its influence on NK cell cytotoxicity. Tumor Biol. 2016, 37, 5455–5466. [Google Scholar] [CrossRef] [PubMed]
  45. Peng, Y.; Xi, C.; Zhu, Y.; Yin, L.; Wei, J.; Zhang, J.-J.; Liu, X.-C.; Guo, S.; Fu, Y.; Miao, Y. Altered expression of CD226 and CD96 on natural killer cells in patients with pancreatic cancer. Oncotarget 2016, 7, 66586–66594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Guillerey, C.; De Andrade, L.F.; Vuckovic, S.; Miles, K.; Ngiow, S.F.; Yong, M.C.R.; Teng, M.W.L.; Colonna, M.; Ritchie, D.S.; Chesi, M.; et al. Immunosurveillance and therapy of multiple myeloma are CD226 dependent. J. Clin. Investig. 2015, 125, 2077–2089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Sivori, S.; Vacca, P.; Del Zotto, G.; Munari, E.; Mingari, M.C.; Moretta, L. Human NK cells: Surface receptors, inhibitory checkpoints, and translational applications. Cell. Mol. Immunol. 2019, 16, 430–441. [Google Scholar] [CrossRef] [PubMed]
  48. Guillamón, C.F.; Martínez-Sánchez, M.V.; Gimeno, L.; Mrowiec, A.; Martínez-García, J.; Server-Pastor, G.; Martínez-Escribano, J.; Torroba, A.; Ferri, B.; Abellán, D.; et al. NK Cell Education in Tumor Immune Surveillance: DNAM-1/KIR Receptor Ratios as Predictive Biomarkers for Solid Tumor Outcome. Cancer Immunol. Res. 2018. [Google Scholar] [CrossRef] [Green Version]
  49. Guillamón, C.F.; Martínez-Sánchez, M.V.; Gimeno, L.; Campillo, J.A.; Server-Pastor, G.; Martínez-García, J.; Martínez-Escribano, J.; Torroba, A.; Ferri, B.; Abellán, D.J.; et al. Activating KIRs on Educated NK Cells Support Downregulation of CD226 and Inefficient Tumor Immunosurveillance. Cancer Immunol. Res. 2019, 7, 1307–1317. [Google Scholar] [CrossRef] [Green Version]
  50. Martínez-Sánchez, M.V.; Periago, A.; Legaz, I.; Gimeno, L.; Mrowiec, A.; Montes-Barqueros, N.R.; Campillo, J.A.; Bolarin, J.M.; Bernardo, M.V.; López-Álvarez, M.R.; et al. Overexpression of KIR inhibitory ligands (HLA-I) determines that immunosurveillance of myeloma depends on diverse and strong NK cell licensing. Oncoimmunology 2016, 5, e1093721. [Google Scholar] [CrossRef]
  51. Gimeno, L.; Martínez-Banaclocha, H.; Bernardo, M.V.; Bolarin, J.M.; Marín, L.; López-Hernández, R.; López-Alvarez, M.R.; Moya-Quiles, M.R.; Muro, M.; Frias-Iniesta, J.F.; et al. NKG2D Polymorphism in Melanoma Patients from Southeastern Spain. Cancers 2019, 11, 438. [Google Scholar] [CrossRef] [Green Version]
  52. Locatelli, F.; Pende, D.; Mingari, M.C.; Bertaina, A.; Falco, M.; Moretta, A.; Moretta, L. Cellular and molecular basis of haploidentical hematopoietic stem cell transplantation in the successful treatment of high-risk leukemias: Role of alloreactive NK cells. Front. Immunol. 2013, 4, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Symons, H.J.; Leffell, M.S.; Rossiter, N.D.; Zahurak, M.; Jones, R.J.; Fuchs, E.J. 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, 533–542. [Google Scholar] [CrossRef] [Green Version]
  54. Nielsen, S.N.; Grell, K.; Nersting, J.; Abrahamsson, J.; Lund, B.; Kanerva, J.; Jónsson, Ó.G.; Vaitkeviciene, G.; Pruunsild, K.; Hjalgrim, L.L.; et al. DNA-thioguanine nucleotide concentration and relapse-free survival during maintenance therapy of childhood acute lymphoblastic leukaemia (NOPHO ALL2008): A prospective substudy of a phase 3 trial. Lancet Oncol. 2017, 18, 515–524. [Google Scholar] [CrossRef]
  55. Ullah, M.A.; Hill, G.R.; Tey, S.-K. Functional Reconstitution of Natural Killer Cells in Allogeneic Hematopoietic Stem Cell Transplantation. Front. Immunol. 2016, 7, 144. [Google Scholar] [CrossRef] [Green Version]
  56. Ek, T.; Mellander, L.; Andersson, B.; Abrahamsson, J. Immune reconstitution after childhood acute lymphoblastic leukemia is most severely affected in the high risk group. Pediatr. Blood Cancer 2005, 44, 461–468. [Google Scholar] [CrossRef]
  57. Shelikhova, L.; Ilushina, M.; Shekhovtsova, Z.; Shasheleva, D.; Khismatullina, R.; Kurnikova, E.; Pershin, D.; Balashov, D.; Radygina, S.; Trakhtman, P.; et al. αβ T Cell-Depleted Haploidentical Hematopoietic Stem Cell Transplantation without Antithymocyte Globulin in Children with Chemorefractory Acute Myelogenous Leukemia. Biol. Blood Marrow Transplant. 2019, 25, e179–e182. [Google Scholar] [CrossRef]
  58. Sharma, A.; Rastogi, N.; Chatterjee, G.; Kapoor, R.; Nivargi, S.; Yadav, S.P. Haploidentical Stem Cell Transplantation With Posttransplant Cyclophosphamide for Pediatric Acute Leukemia is Safe and Effective. J. Pediatr. Hematol. Oncol. 2020. [Google Scholar] [CrossRef] [PubMed]
  59. Katsanis, E.; Sapp, L.N.; Reid, S.C.; Reddivalla, N.; Stea, B. T-Cell Replete Myeloablative Haploidentical Bone Marrow Transplantation Is an Effective Option for Pediatric and Young Adult Patients With High-Risk Hematologic Malignancies. Front. Pediatr. 2020, 8, 282. [Google Scholar] [CrossRef] [PubMed]
  60. Schmiegelow, K. Maintenance therapy of childhood acute lymphoblastic leukemia: Do all roads lead to Rome? Pediatr. Blood Cancer 2020, 67, e28418. [Google Scholar] [CrossRef] [PubMed]
  61. Li, B.; Brady, S.W.; Ma, X.; Shen, S.; Zhang, Y.; Li, Y.; Szlachta, K.; Dong, L.; Liu, Y.; Yang, F.; et al. Therapy-induced mutations drive the genomic landscape of relapsed acute lymphoblastic leukemia. Blood 2020, 135, 41–55. [Google Scholar] [CrossRef] [PubMed]
  62. Apps, R.; Carrington, M. Response to Comment on “Influence of HLA-C Expression Level on HIV Control. Science 2013, 341, 1175. [Google Scholar] [CrossRef] [Green Version]
  63. Babor, F.; Manser, A.R.; Fischer, J.C.; Scherenschlich, N.; Enczmann, J.; Chazara, O.; Moffett, A.; Borkhardt, A.; Meisel, R.; Uhrberg, M. KIR ligand C2 is associated with increased susceptibility to childhood ALL and confers an elevated risk for late relapse. Blood 2014, 124, 2248–2251. [Google Scholar] [CrossRef]
  64. Bortin, M.M.; D’Amaro, J.; Bach, F.H.; Rimm, A.A.; van Rood, J.J. HLA associations with leukemia. Blood 1987, 70, 227–232. [Google Scholar] [CrossRef]
  65. Caruso, C.; Cammarata, G.; Sireci, G.; Modica, M.A. HLA-Cw4 association with acute lymphoblastic leukaemia in Sicilian patients. Vox Sang. 1988, 54, 57–58. [Google Scholar]
  66. Baldauf, C.; Kubel, M. [HLA and leukemia]. Folia Haematol. Int. Mag. Klin. Morphol. Blutforsch. 1989, 116, 123–132. [Google Scholar]
  67. Karabon, L.; Jedynak, A.; Giebel, S.; Wołowiec, D.; Kielbinski, M.; Woszczyk, D.; Kapelko-Slowik, K.; Kuliczkowski, K.; Frydecka, I. KIR/HLA gene combinations influence susceptibility to B-cell chronic lymphocytic leukemia and the clinical course of disease. Tissue Antigens 2011, 78, 129–138. [Google Scholar] [CrossRef]
  68. Arima, N.; Kanda, J.; Tanaka, J.; Yabe, T.; Morishima, Y.; Kim, S.-W.; Najima, Y.; Ozawa, Y.; Eto, T.; Kanamori, H.; et al. Homozygous HLA-C1 is Associated with Reduced Risk of Relapse after HLA-Matched Transplantation in Patients with Myeloid Leukemia. Biol. Blood Marrow Transplant. 2018, 24, 717–725. [Google Scholar] [PubMed] [Green Version]
  69. Cárceles-Álvarez, A.; Ortega-García, J.A.; López-Hernández, F.A.; Fuster-Soler, J.L.; Ramis, R.; Kloosterman, N.; Castillo, L.; Sánchez-Solís, M.; Claudio, L.; Ferris-Tortajada, J. Secondhand smoke: A new and modifiable prognostic factor in childhood acute lymphoblastic leukemias. Environ. Res. 2019, 178, 108689. [Google Scholar] [CrossRef]
  70. Mastaglio, S.; Wong, E.; Perera, T.; Ripley, J.; Blombery, P.; Smyth, M.J.; Koldej, R.; Ritchie, D. Natural killer receptor ligand expression on acute myeloid leukemia impacts survival and relapse after chemotherapy. Blood Adv. 2018, 2, 335–346. [Google Scholar] [PubMed] [Green Version]
  71. Epling-Burnette, P.K.; Bai, F.; Painter, J.S.; Rollison, D.E.; Salih, H.R.; Krusch, M.; Zou, J.; Ku, E.; Zhong, B.; Boulware, D.; et al. Reduced natural killer (NK) function associated with high-risk myelodysplastic syndrome (MDS) and reduced expression of activating NK receptors. Blood 2007, 109, 4816–4824. [Google Scholar] [CrossRef] [PubMed]
  72. Sabattini, E.; Bacci, F.; Sagramoso, C.; Pileri, S.A. WHO classification of tumours of haematopoietic and lymphoid tissues in 2008: An overview. Pathologica 2010, 102, 83–87. [Google Scholar]
Figure 1. Patient outcome according to the type of acute leukemia and the risk status at enrollment. (A) Kaplan–Meier and log-rank tests for cumulative incidence of relapse, treatment-related mortality, event-free survival (EFS), and overall survival (OS) according to the type of acute leukemia (B-ALL, T-ALL, or AML) and the risk status at enrollment. (B) Rates (%) of cumulative incidence of relapse, treatment-related mortality, EFS, and OS according to the risk status of patients at diagnosis for each type of leukemia. Primary data can be consulted in File S1.
Figure 1. Patient outcome according to the type of acute leukemia and the risk status at enrollment. (A) Kaplan–Meier and log-rank tests for cumulative incidence of relapse, treatment-related mortality, event-free survival (EFS), and overall survival (OS) according to the type of acute leukemia (B-ALL, T-ALL, or AML) and the risk status at enrollment. (B) Rates (%) of cumulative incidence of relapse, treatment-related mortality, EFS, and OS according to the risk status of patients at diagnosis for each type of leukemia. Primary data can be consulted in File S1.
Cancers 13 02294 g001
Figure 2. Expression of ligands for NK cell receptors on leukemic blast cells. (A) Mean fluorescence intensity (MFI) expression on normal counterpart cells (control, striped bars) and leukemic blast cells (solid bars) of ligands for activating NK cell receptors MICA/B and ULBP (ligands of NKG2D) and CD112 and CD155 (ligands of DNAM-1/CD226). (B) Expression of HLA-I and HLA-C ligands for inhibitory receptors (iKIRs and NKG2A). See Figure 7 for details of MFI estimation in leukocyte subset.
Figure 2. Expression of ligands for NK cell receptors on leukemic blast cells. (A) Mean fluorescence intensity (MFI) expression on normal counterpart cells (control, striped bars) and leukemic blast cells (solid bars) of ligands for activating NK cell receptors MICA/B and ULBP (ligands of NKG2D) and CD112 and CD155 (ligands of DNAM-1/CD226). (B) Expression of HLA-I and HLA-C ligands for inhibitory receptors (iKIRs and NKG2A). See Figure 7 for details of MFI estimation in leukocyte subset.
Cancers 13 02294 g002
Figure 3. Event-free survival (EFS) of patients according to the expression of ligands for NK cell receptors. (A) Frequency of HLA-I and HLA-C loss in B-ALL, T-ALL and AML patients. HLA loss was computed when the mean fluorescence intensity (MFI) expression of HLA on leukemic blast cells was lower than that observed on the normal counterpart cells. P estimated with the Chi-squared test. (B) Kaplan–Meier and log-rank tests for EFS of acute leukemia patients according to the low or high expression of ligands for activating NK cell receptors MICA/B (ROC, AUC = 0.53, cutoff = 108.5, sensitivity = 80.0, and specificity = 51.0), ULBP-1 (ROC, AUC = 0.60, cutoff = 140.1, sensitivity = 86.6%, and specificity = 60.1%), CD112 (ROC, AUC = 0.55, cutoff = 1640, sensitivity = 66.0%, and specificity = 55.0%) and CD155 (ROC, AUC = 0.54, cutoff = 326.1, sensitivity = 56.0, and specificity = 62.2). (C) Kaplan–Meier and log-rank tests for EFS of acute leukemia patients according to the loss or preservation of HLA-I and HLA-C.
Figure 3. Event-free survival (EFS) of patients according to the expression of ligands for NK cell receptors. (A) Frequency of HLA-I and HLA-C loss in B-ALL, T-ALL and AML patients. HLA loss was computed when the mean fluorescence intensity (MFI) expression of HLA on leukemic blast cells was lower than that observed on the normal counterpart cells. P estimated with the Chi-squared test. (B) Kaplan–Meier and log-rank tests for EFS of acute leukemia patients according to the low or high expression of ligands for activating NK cell receptors MICA/B (ROC, AUC = 0.53, cutoff = 108.5, sensitivity = 80.0, and specificity = 51.0), ULBP-1 (ROC, AUC = 0.60, cutoff = 140.1, sensitivity = 86.6%, and specificity = 60.1%), CD112 (ROC, AUC = 0.55, cutoff = 1640, sensitivity = 66.0%, and specificity = 55.0%) and CD155 (ROC, AUC = 0.54, cutoff = 326.1, sensitivity = 56.0, and specificity = 62.2). (C) Kaplan–Meier and log-rank tests for EFS of acute leukemia patients according to the loss or preservation of HLA-I and HLA-C.
Cancers 13 02294 g003
Figure 4. KIR2DL1/HLA-C2-epitope and specifically KIR2DL1/HLA-C*04 interactions are associated with reduced EFS and OS of acute leukemic pediatric patients. (A) Five-year event-free survival (5y-EFS) rates for KIR/HLA-ligand interaction (left) and for KIR/HLA-C allotypes with the C2-epitope (right). (B) Kaplan–Meier and log-rank tests for EFS of total, B-ALL, T-ALL and AML patients according to the presence of KIR2DL1/C2-epitope interaction. (C) Kaplan–Meier and log-rank tests for EFS and OS of acute leukemia patients according to the presence of KIR2DL1/HLA-C*04 interaction. (D) Kaplan–Meier and log-rank tests for EFS of acute leukemia patients according to the presence of KIR2DL1/HLA-C*05 interaction (left) or according to the combined presence of KIR2DL1/HLA-C*04 and KIR2DL1/HLA-C*05 interactions (right).
Figure 4. KIR2DL1/HLA-C2-epitope and specifically KIR2DL1/HLA-C*04 interactions are associated with reduced EFS and OS of acute leukemic pediatric patients. (A) Five-year event-free survival (5y-EFS) rates for KIR/HLA-ligand interaction (left) and for KIR/HLA-C allotypes with the C2-epitope (right). (B) Kaplan–Meier and log-rank tests for EFS of total, B-ALL, T-ALL and AML patients according to the presence of KIR2DL1/C2-epitope interaction. (C) Kaplan–Meier and log-rank tests for EFS and OS of acute leukemia patients according to the presence of KIR2DL1/HLA-C*04 interaction. (D) Kaplan–Meier and log-rank tests for EFS of acute leukemia patients according to the presence of KIR2DL1/HLA-C*05 interaction (left) or according to the combined presence of KIR2DL1/HLA-C*04 and KIR2DL1/HLA-C*05 interactions (right).
Cancers 13 02294 g004
Figure 5. Loss of HLA-C expression was selectively observed in patients carrying the HLA-C*04 allotype. HLA-C and HLA-I expression on leukemic blast cells and on control cells in total acute leukemia patients and B-ALL patients carrying HLA-C allotypes with the C2-epitope. p estimated with Student’s t-test.
Figure 5. Loss of HLA-C expression was selectively observed in patients carrying the HLA-C*04 allotype. HLA-C and HLA-I expression on leukemic blast cells and on control cells in total acute leukemia patients and B-ALL patients carrying HLA-C allotypes with the C2-epitope. p estimated with Student’s t-test.
Cancers 13 02294 g005
Figure 6. KIR2DL1/C*04 interaction is an independent prognostic biomarker. (A) Cox regression analysis for PFS and OS for sex, age, type of leukemia, risk status at enrollment, low/high expression of ULBP-1, and presence/absence of the KIR2DL1/C*04 interaction. (B) Kaplan–Meier and log-rank tests for EFS according to the presence/absence of KIR2DL1/HLA-C*04 interaction in standard/intermediate and high-risk patients. (C) Kaplan–Meier and log-rank tests for OS according to the presence/absence of KIR2DL1/HLA-C*04 interaction in patients enrolled because of new diagnosis or relapse.
Figure 6. KIR2DL1/C*04 interaction is an independent prognostic biomarker. (A) Cox regression analysis for PFS and OS for sex, age, type of leukemia, risk status at enrollment, low/high expression of ULBP-1, and presence/absence of the KIR2DL1/C*04 interaction. (B) Kaplan–Meier and log-rank tests for EFS according to the presence/absence of KIR2DL1/HLA-C*04 interaction in standard/intermediate and high-risk patients. (C) Kaplan–Meier and log-rank tests for OS according to the presence/absence of KIR2DL1/HLA-C*04 interaction in patients enrolled because of new diagnosis or relapse.
Cancers 13 02294 g006
Figure 7. Immunophenotype analysis of NK cell receptor ligands in normal and leukemic bone marrow cells. (A) Events with low/intermediate dispersion in FSC/SSC, including residual normal lymphocytes (brown dots) and leukemic blast cells (blue dots), were gated in an FSC/SSC dot plot. Leukemic cells were gated in a plot with CD45-APCCy7 combined with CD19-PECy7 for B-ALL, CD7-PECy7 for T-ALL or CD34-PECy7 or CD33-PECy7 for AML. Granulocytes (green dots) were identified with gates on high SCC in the FSC/SSC dot plot and low CD45 in the CD45-APCCy7/PECy7 dot plot. Mean fluorescence intensity (MFI) was evaluated for CD112, CD155, and ULBP-1 in the PE-channel, for MICA/B in the APC-channel, for HLA-I in the FITC-channel, and for HLA-C in the V450 (VB421) channel on lymphocytes, granulocytes, and blast cells. Dot-plots correspond to a representative case with a clear expression loss of HLA-I and HLA-C and strong gain of CD112, with a moderate gain of MICA/B, ULBP-1, and CD155. (B) MFI in the FITC, PE, APC, and V450 (BV421) channels were also estimated for leukocyte subsets in the isotype tube to determine the background of each fluorescence.
Figure 7. Immunophenotype analysis of NK cell receptor ligands in normal and leukemic bone marrow cells. (A) Events with low/intermediate dispersion in FSC/SSC, including residual normal lymphocytes (brown dots) and leukemic blast cells (blue dots), were gated in an FSC/SSC dot plot. Leukemic cells were gated in a plot with CD45-APCCy7 combined with CD19-PECy7 for B-ALL, CD7-PECy7 for T-ALL or CD34-PECy7 or CD33-PECy7 for AML. Granulocytes (green dots) were identified with gates on high SCC in the FSC/SSC dot plot and low CD45 in the CD45-APCCy7/PECy7 dot plot. Mean fluorescence intensity (MFI) was evaluated for CD112, CD155, and ULBP-1 in the PE-channel, for MICA/B in the APC-channel, for HLA-I in the FITC-channel, and for HLA-C in the V450 (VB421) channel on lymphocytes, granulocytes, and blast cells. Dot-plots correspond to a representative case with a clear expression loss of HLA-I and HLA-C and strong gain of CD112, with a moderate gain of MICA/B, ULBP-1, and CD155. (B) MFI in the FITC, PE, APC, and V450 (BV421) channels were also estimated for leukocyte subsets in the isotype tube to determine the background of each fluorescence.
Cancers 13 02294 g007
Table 1. Biological, clinical, therapeutic, and evolutionary features of patients.
Table 1. Biological, clinical, therapeutic, and evolutionary features of patients.
Total
(n = 102)
B-ALL
(n = 70)
T-ALL
(n = 16)
AML
(n = 16)
Age (month, mean ± SD)83.0 ± 49.382.5 ± 49.191.7 ± 44.976.6 ± 55.6
Sex (male, n (%))63 (61.8%)42 (60.0%)12 (75.0%)9 (56.3%)
Inclusion in the study
At diagnosis, n (%)96 (94.1%)66 (94.3%)16 (100%)14 (87.5%)
At relapse, n (%)6 (5.9%)4 (5.7%) 2 (12.5%)
PB-WBC count at diagnosis
<10 × 103/µL89 (87.3%)65 (92.9%)10 (62.5%)14 (87.5%)
10–50 × 103/µL5 (4.9%)4 (5.7%) 1 (6.3%)
>50 × 103/µL8 (7.8%)1 (1.4%)6 (37.5%)1 (6.3%)
Risk stratification 1
Standard, n (%)27 (26.4%)23 (32.9%)0 (0.0%)3 (18.7%)
Intermediate, n (%)41 (40.2%) 29 (41.1%)11 (68.7%)2 (12.5%)
High, n (%)34 (34.0%)18 (25.7%)5 (31.3%)11 (68.8%)
Treatments
PETHEMA-96/2001/200513 (12.6%)12 (17.0%)1 (6.2%)
LAL-SEHOP-PETHEMA-201373 (71.4%)58 (82.7%)15 (93.7%)
LMA-SHOP-200710 (9.8%) 10 (62.5%)
NOPHO-DBH-AML-20126 (5.8%) 6 (37.5%)
HSCT, n (%)18 (17.6%)8 (11.4%)3 (18.8%)7 (43.8%)
Allo-HSCT, n (%)13 (12.7%)5 (7.1%)3 (18.8%)5 (31.3%)
Related, n (%)5 (38.5%)2 (40.0%)2 (66.7%)3 (42.9%)
Unrelated, n (%)8 (61.5%)3 (60.0%)1 (33.3%)4 (57.1%)
Haplo-HSCT, n (%)5 (4.9%)3 (4.3%) 2 (12.5%)
Clinical outcome
Mean follow-up (months)41.98 ± 31.541.9 ± 32.946.6 ± 30.637.7 ± 25.9
Relapse, n (%)9 (8.8%)2 (2.8%)2 (12.5%)5 (31.3%)
Death, n (%)12 (11.8%)7 (10.0%) 5 (31.3%)
1 Risk stratification: according to specific criteria (described in method section); B-ALL: B-cell acute lymphoblastic leukemia; T-ALL: T-cell acute lymphoblastic leukemia; AML: acute myeloid leukemia; SD: standard deviation; BM: bone marrow; PB: peripheral blood; HSCT: hematopoietic stem cell transplantation; EFS: event-free survival; OS: overall survival; control group (n = 83): age 74.4 ± 35.7 months. Male n = 47 (56.6%); female n = 36 (43.4%).
Table 2. KIR gene, HLA-I ligand, and KIR/HLA-I ligand interaction frequencies in controls and patients.
Table 2. KIR gene, HLA-I ligand, and KIR/HLA-I ligand interaction frequencies in controls and patients.
KIR Genes, n (%)Controls
(n = 83)
Patients
(n = 102)
B-ALL
(n = 70)
T-ALL
(n = 16)
AML
(n = 16)
KIR2DL180 (96.4%)99 (97.1%)68 (97.1%)16 (100%)15 (93.8%)
KIR2DL2/2DS2 148 (57.8%)49 (48.0%)32 (45.7%)7 (43.8%)10 (62.5%)
KIR2DL374 (89.2%)93 (91.2%)64 (91.4%)15 (93.8%)14 (87.5%)
KIR2DL544 (53.0%)56 (54.9%)35 (50.0%)8 (50.0%)13 (81.3%)
KIR3DL181 (97.6%)100 (98.0%)68 (97.1%)16 (100%)16 (100%)
KIR2DS130 (36.1%)38 (37.3%)23 (32.9%)7 (43.8%)8 (50.0%)
KIR2DS326 (31.3%)35 (34.3%)21 (30.0%)5 (31.3%)9 (56.3%)
KIR2DS4full35 (42.2%)54 (52.9%)34 (48.6%)11 (68.8%)9 (56.3%)
KIR2DS526 (31.3%)30 (29.4%)19 (27.1%)6 (37.5%)5 (31.3%)
KIR3DS129 (34.9%)38 (37.3%)24 (34.3%)7 (43.8%)7 (43.8%)
HLA-I ligands, n (%)
HLA-A*03/A*1123 (27.7%)30 (30.6%)20 (30.8%)4 (25.0%)6 (37.5%)
Bw4 epitope65 (78.3%)70 (68.6%)49 (70.0%)10 (62.5%)11 (68.8%)
Bw4-80T20 (24.1%)22 (21.6%)16 (22.9%)4 (25.0%)2 (12.5%)
Bw4-80I53 (63.9%)61 (59.8%)44 (62.9%)8 (50.0%)9 (56.3.3%)
HLA-C1 epitope61 (73.5%)80 (78.4%)56 (80.0%)13 (81.3%)11 (68.8%)
HLA-C2 epitope56 (67.5%)73 (71.6%)48 (68.6%)10 (62.5%)15 (93.8%)
KIR2DS4-ligands 244 (53.0%)71 (71.0%) 349 (72.1%)10 (62.5%)12 (75.0%)
KIR/HLA-ligand interactions, n (%)
KIR2DL1/C254 (65.1%)70 (68.6%)46 (65.7%)10 (62.5%)14 (87.5%)
KIR2DL2/C134 (41.0%)38 (37.3%)27 (38.6%)5 (31.3%)6 (37.5%)
KIR2DL3/C154 (65.1%)73 (71.6%)52 (74.3%)12 (75.0%)9 (56.3%)
KIR3DL1/Bw463 (75.9%)66 (68.0%)45 (69.2%)10 (62.5%)11 (68.8%)
KIR2DS1/C222 (26.5%)28 (27.5%)16 (22.9%)5 (31.3%)7 (43.8%)
KIR2DS4/Ligands 118 (21.7%)33 (35.1%)23 (35.4%)5 (31.1%)6 (37.5%)
KIR2DS4/C*04:017 (8.4%)15 (15.5%)10 (15.4%)2 (15.5%)3 (18.8%)
KIR3DS1/B*513 (3.6%)4 (3.92%)2 (2.85%)2 (12.5%)0 (0.0%)
1 KIR2DL2 and KIR2DS2 had a high linkage disequilibrium and showed the same frequency; 2 ligands for KIR2DS4: HLA-A*11, C*01:02, C*02:02, C*04:01, C*05:01, C*14:02, and C*16:01 [52]. 3 Controls vs. patients p = 0.009, Pc = 0.045.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martínez-Sánchez, M.V.; Fuster, J.L.; Campillo, J.A.; Galera, A.M.; Bermúdez-Cortés, M.; Llinares, M.E.; Ramos-Elbal, E.; Pascual-Gázquez, J.F.; Fita, A.M.; Martínez-Banaclocha, H.; et al. Expression of NK Cell Receptor Ligands on Leukemic Cells Is Associated with the Outcome of Childhood Acute Leukemia. Cancers 2021, 13, 2294. https://doi.org/10.3390/cancers13102294

AMA Style

Martínez-Sánchez MV, Fuster JL, Campillo JA, Galera AM, Bermúdez-Cortés M, Llinares ME, Ramos-Elbal E, Pascual-Gázquez JF, Fita AM, Martínez-Banaclocha H, et al. Expression of NK Cell Receptor Ligands on Leukemic Cells Is Associated with the Outcome of Childhood Acute Leukemia. Cancers. 2021; 13(10):2294. https://doi.org/10.3390/cancers13102294

Chicago/Turabian Style

Martínez-Sánchez, María Victoria, José Luis Fuster, José Antonio Campillo, Ana María Galera, Mar Bermúdez-Cortés, María Esther Llinares, Eduardo Ramos-Elbal, Juan Francisco Pascual-Gázquez, Ana María Fita, Helios Martínez-Banaclocha, and et al. 2021. "Expression of NK Cell Receptor Ligands on Leukemic Cells Is Associated with the Outcome of Childhood Acute Leukemia" Cancers 13, no. 10: 2294. https://doi.org/10.3390/cancers13102294

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop