Natural killer (NK) cells play an important role in surveillance and elimination of malignant cells. Their spontaneous cytotoxicity was first demonstrated in vitro against leukemia cell lines, and NK cells might play a crucial role in the therapy of leukemia. NK cell activity is controlled by an array of germ line–encoded activating and inhibitory receptors, as well as modulating coreceptors. This biologic feature can be exploited in allogeneic cell therapy, and the recognition of “missing-self” on target cells is crucial for promoting NK cell–mediated graft-versus-leukemia effects. In this regard, NK cells that express an inhibitory killer immunoglobulin-like receptor (iKIR) for which the respective major histocompatibility complex class I ligand is absent on leukemic target cells can exert alloreactivity in vitro and in vivo. Several models regarding potential donor–patient constellations have been described that have demonstrated the clinical benefit of such alloreactivity of the donor-derived NK cell system in patients with adult acute myeloid leukemia and pediatric B-cell precursor acute lymphoblastic leukemia after allogeneic stem cell transplantation. Moreover, adoptive transfer of mature allogeneic NK cells in the nontransplant or transplant setting has been shown to be safe and feasible, whereas its effectivity needs further evaluation. NK cell therapy can be further improved by optimal donor selection based on phenotypic and genotypic properties, by adoptive transfer of NK cells with ex vivo or in vivo cytokine stimulation, by the use of antibodies to induce antibody-dependent cellular cytotoxicity or to block iKIRs, or by transduction of chimeric antigen receptors.

Natural killer (NK) cells belong to innate lymphoid immune cells that contribute to antitumor responses without prior sensitization. According to the currently applied innate lymphoid cell nomenclature, NK cells are a prototypical member of the group 1 innate lymphoid cells insofar as they produce interferon γ (IFN-γ) and developmentally require the common cytokine receptor γ chain, the transcriptional repressor inhibitor of DNA binding 2, and T-bet.1  During maturation, NK cells sequentially acquire lineage-specific markers such as CD94, NKp46, CD56, and CD16. Based on the surface density of CD56, the primarily cytokine-producing CD56brightCD16 NK cell subset is distinguishable from the predominantly cytotoxic CD56dimCD16+ subset. Although CD56brightCD16 NK cells constitute a minor fraction of NK cells in the peripheral blood (∼10%), they are enriched in the secondary lymphoid organs and presumably differentiate here to CD56dimCD16+ NK cells. During this maturational process, they upregulate natural cytotoxicity receptors, express perforin, proliferate, and acquire cytolytic activity upon stimulation with the T cell–derived cytokine interleukin 2 (IL-2). In contrast, the majority (∼90%) of the peripheral blood NK cell compartment consists of CD56dimCD16+ NK cells, which display markers of late maturity such as killer immunoglobulin-like receptors (KIRs) that are inhibitory (iKIRs) and CD57, vividly produce perforin to exert effector functions, but display little proliferative capacity upon IL-2 stimulation.2 

NK cells use different innate receptors to sense their environment and respond to alterations induced by infections or by malignant cell transformation. In a process termed “licensing,” NK cells use iKIRs for “self”–major histocompatibility complex (MHC) class I molecules to maintain a state of responsiveness and to kill target cells that have lost MHC class I.3  In a balanced state, healthy tissue that expresses the self ligands to inhibitory NK cell receptors (ie, certain MHC class I molecules) will be spared from being killed. In contrast, the recognition of missing or downregulated self-MHC class I molecules on tumor cells by licensed NK cells shifts the NK cell receptor balance toward activation. Altogether, the modulation of NK cell activity is controlled by an array of germ line–encoded activating and inhibitory receptors, as well as modulating coreceptors, which are reviewed by Long et al4  and summarized in Figure 1.

Figure 1

Overview over important activatory, inhibitory, and costimulatory NK cell receptors. NK cells integrate various signals in response to leukemic cells. The precise function of some of the receptors is not yet well known, and some activatory receptors might be costimulatory, and vice versa. Note that recent evidence suggests that NKG2D and NKp46 are co-stimulatory receptors because they are not able to induce NK cell activity on their own.4  In addition, KIR2DL4 is, to date, considered to convey activatory rather than inhibitory signals.4  Many other receptors, including cytokine, chemotactic, and adhesion molecule receptors, as well as other costimulatory receptors, are not shown. Ligands are shown in parentheses. AICL; activation-induced C-type lectin; BAT-3, HLA-B-associated transcript 3; CD16, FcRIII receptor; LFA-1, leukocyte functional antigen 1; MICA/B, MHC class I–related chain A/B; NTB, natural killer T- and B-cell antigen; PCNA, proliferating cell nuclear antigen; ULBPs, UL-16 binding proteins.

Figure 1

Overview over important activatory, inhibitory, and costimulatory NK cell receptors. NK cells integrate various signals in response to leukemic cells. The precise function of some of the receptors is not yet well known, and some activatory receptors might be costimulatory, and vice versa. Note that recent evidence suggests that NKG2D and NKp46 are co-stimulatory receptors because they are not able to induce NK cell activity on their own.4  In addition, KIR2DL4 is, to date, considered to convey activatory rather than inhibitory signals.4  Many other receptors, including cytokine, chemotactic, and adhesion molecule receptors, as well as other costimulatory receptors, are not shown. Ligands are shown in parentheses. AICL; activation-induced C-type lectin; BAT-3, HLA-B-associated transcript 3; CD16, FcRIII receptor; LFA-1, leukocyte functional antigen 1; MICA/B, MHC class I–related chain A/B; NTB, natural killer T- and B-cell antigen; PCNA, proliferating cell nuclear antigen; ULBPs, UL-16 binding proteins.

Close modal

Because attempts to exploit autologous NK cells by either in vivo stimulation with IL-25  or the infusion of ex vivo stimulated autologous NK cells6  showed limited clinical efficacy, the focus of research was directed toward the use of NK cells from healthy related or unrelated donors in the context of allogeneic cell therapy.

In this context, donor NK cells that express an iKIR for which the respective ligand is absent on the recipient’s leukemic cells and that concomitantly lack the expression of CD94/NKG2A (because HLA-E molecules are present on all cells and will, as a result, inhibit NK cell functionality) are defined as being alloreactive.7  KIRs are genetically determined and highly polymorphic receptors that recognize allotypic variants of MHC class I alleles (KIR ligands [KIRLs]).8  Molecular cloning has revealed that distinct groups of family members comprise 2 or 3 extracellular immunoglobulin-like domains and, hence, are designated KIR2D and KIR3D, respectively. KIRs with inhibitory activity are long-tailed in their intracytoplasmic region, as indicated by the letter “L” (ie, 2DL1), whereas their activating KIR (aKIR) counterparts are short-tailed, as indicated by the letter “S” (ie, 2DS1). KIR2D family members recognize HLA-C alleles with Lys80 (C2 epitope) or Asn80 (C1 epitope) residues, whereas KIR3D family members recognize HLA-B alleles with a Bw4 supertypic specificity.9  Based on the analysis of the 17 KIR genes and pseudogenes, 2 donor haplotypes are distinguished. KIR A haplotype donors express a canonical gene content, including 6 iKIRs (2DL1, -3, and -4 and 3DL1, -2, and -3) and the activating KIR2DS4, whereas KIR B haplotype donors have a variable gene content and express, in addition to the above-mentioned iKIRs, 1 or more of the B-specific genes: KIR2DS1, -2, -3, -5, KIR2DL2 and/or KIR2DL5.10  The considerable differences that exist in KIR gene content and copy number, together with the extensive allelic polymorphisms, account for the high variability that exists in the genomic KIR region of 2 different individuals.11  Because HLA and KIR segregate independently on different chromosomes and because the expression of KIR genes follows a random stochastic distribution, the repertoire of NK cells with diverging combinations of KIRs is large, and only a minority of HLA-matched donors will also be KIR-KIRL matched.

The beneficial effect of a KIR-KIRL mismatched donor–recipient constellation in leukemia has been initially described by the Perugia group.12,13  In pioneering studies, these researchers provided evidence in adult acute myeloid leukemia (AML), but not in adult B-cell precursor (BCP) acute lymphoblastic leukemia (ALL), that transplantation of grafts from KIR-KIRL mismatched donors enhanced survival rates. This graft-versus-leukemia (GVL) effect was attributed to NK cell alloreactivity and predicted by analysis of the HLA types of donor and recipient.12,13  Since then, a number of clinical studies confirmed these observations,14-16  whereas other studies were not able to demonstrate any benefit from selecting a KIR-KIRL mismatched donor when transplanting AML patients.17-21  To explain these potential differences and to dichotomize the relative risk of relapse as a result of a given KIR-KIRL constellation, various models have been proposed. One is the “ligand–ligand” model suggested by the Perugia group, which predicts the relative risk of relapse, considering mismatches between donor and recipient iKIRLs (ie, HLA disparities in graft-versus-host disease [GVHD] direction).12,22  Others failed to demonstrate improved survival when applying this model and performing haploidentical transplants with less-vigorous modes of T-cell depletion.23-25  A second model, the “receptor–ligand” model suggested by the Memphis group,26,27  considers the presence of iKIRs on donor NK cells, together with the absence of the corresponding KIRL, in the recipient’s HLA repertoire. This model, so far, has been the most accurate when performing correlative analyses between the donor KIR phenotype and the risk of relapse in pediatric BCP-ALL patients,26,27  in T cell–depleted matched sibling transplants,28  and in unrelated donor transplants of adult AML patients.29,30  The less commonly applied “gene–gene” model accounts for mismatches on donor and recipient KIR genes31  and has accurately predicted survival rates in KIR haplotype B donors after nonmyeloablative hematopoietic stem cell transplantation (HSCT).32  The Tuebingen group33,34  observed that patients homozygous for a HLA-C1 alleles have a poorer outcome than other patients. As a result of these conceptual differences, the term “KIR-KIRL mismatch” is used inconsistently, and this inconsistency may explain, in part, why some of the models are more or less able to predict outcome. In Figure 2, possible constellations of alloreactivity are shown.

Figure 2

Possible constellations of NK cell alloreactivity are depicted. (A) Licensed donor NK cells (ie, NK cells that have iKIRs for self–HLA class I) are inhibited via engagement of the iKIR by the recipients’ ligands (HLA class I), which exert a strong inhibitory signal; thus, these donor NK cells cannot lyse recipients’ leukemic blasts (NK cell nonalloreactivity). (B) iKIRs of licensed NK cells are not engaged by the KIRLs, and the donor NK cells are activated and lyse recipients’ leukemic blasts (NK cell alloreactivity). (C) Recipients’ blasts lack HLA class I expression and, therefore, cannot inhibit donor NK cells, resulting in activation of donor NK cells. (D) NK cells do not express KIRs and, therefore, have not been licensed. KIR-negative NK cells are one of the earliest lymphocyte populations that reconstitute after T cell–depleted HSCT. These NK cells are hyporesponsive but might become responsive upon cytokine stimulation.

Figure 2

Possible constellations of NK cell alloreactivity are depicted. (A) Licensed donor NK cells (ie, NK cells that have iKIRs for self–HLA class I) are inhibited via engagement of the iKIR by the recipients’ ligands (HLA class I), which exert a strong inhibitory signal; thus, these donor NK cells cannot lyse recipients’ leukemic blasts (NK cell nonalloreactivity). (B) iKIRs of licensed NK cells are not engaged by the KIRLs, and the donor NK cells are activated and lyse recipients’ leukemic blasts (NK cell alloreactivity). (C) Recipients’ blasts lack HLA class I expression and, therefore, cannot inhibit donor NK cells, resulting in activation of donor NK cells. (D) NK cells do not express KIRs and, therefore, have not been licensed. KIR-negative NK cells are one of the earliest lymphocyte populations that reconstitute after T cell–depleted HSCT. These NK cells are hyporesponsive but might become responsive upon cytokine stimulation.

Close modal

Recently emerging evidence indicates that not only the presence of iKIRs but also the presence of the activating KIR2DS1 regulates NK cell cytotoxicity against lymphoblastoid cell lines,35  adult AML and BCP-ALL,36-39  and pediatric AML and BCP-ALL.40,41  The inclusion of KIR haplotype variability has added an additional layer of complexity to the question of which donor will be the “optimal” donor. Elegant studies in adult AML patients demonstrated that the selection of an HLA-matched unrelated donor who possesses aKIRs next to iKIRs will confer a significantly higher event-free survival.36-38  Depending on their exact position on the KIR locus, distinct centromeric (Cen) and telomeric (Tel) gene-content motifs are described in KIR haplotype A and B donors. In a large cohort study, Cen- and Tel-B motifs both contributed to relapse protection, but Cen-B homozygosity had the strongest independent protective effect.37  This was particularly true for recipients with 1 or 2 C1-bearing HLA-C allotypes.42  In an attempt to calculate the relative risk of relapse after transplant, the so-called KIR B content score can be calculated (http://www.ebi.ac.uk/ipd/kir/donor_b_content.html) by scoring the number of Cen-B and/or Tel-B motifs in each genotype, thus allowing the grouping of donors into the categories of neutral, better, and best. In a large adult cohort study, matched unrelated donors with a KIR B content score of 2 or more conferred a distinctly higher event-free survival in patients with AML, but not ALL.37  A similar result was obtained in children with BCP-ALL after haploidentical T cell–depleted transplantation: not only the presence of KIR haplotype B but, even more so, the selection of donors with a high KIR B content score >2 conferred better protection against relapse.43  In conclusion, a number of association studies suggest that a donor with an iKIR-KIRL mismatch toward the recipient who possesses aKIRs next to iKIRs and who, ideally, expresses not 1, but multiple, aKIRs will be a potentially optimal donor that may promote clinically relevant NK cell alloreactivity (Figure 3). However, given the complexity of the transplant procedure and the strong linkage disequilibrium of selected aKIR genes to other KIR genes, further clinical studies incorporating functional NK cell analysis are needed to conclusively answer the question of whether aKIRs directly contribute to NK cell alloreactivity.

Figure 3

Defining the optimal donor. Based on the KIR haplotype A that has a fixed gene content of 6 iKIRs and 1 aKIR, and the haplotype B that has a highly variable gene content and up to 5 aKIRs, donors can be assigned either to KIR genotype A/A (ie, homozygous for A haplotypes) or to B/x (having 1 or 2 B haplotypes). The Cen and Tel regions can reassemble to form recombinant haplotypes. Genetic association studies suggest that an optimal alloreactive donor will be one with an iKIR-KIRL mismatch and who possesses multiple aKIRs (haplotype B) next to iKIRs. The KIR B content score mainly reflects the number of aKIRs, and the highest score is associated with the highest number of aKIRs. NK cells from such donors with a high KIR B content score should, in principle, exert strong alloreactive antileukemic NK cell activity, and it has been shown that transplantation with donors homozygous for Cen KIR B haplotypes is associated with the lowest level of relapse and highest overall survival.36,42,43  The KIR B content score can easily be determined by KIR genotyping. Note that the majority of haplotype B donors do not express all genes shown in the figure.

Figure 3

Defining the optimal donor. Based on the KIR haplotype A that has a fixed gene content of 6 iKIRs and 1 aKIR, and the haplotype B that has a highly variable gene content and up to 5 aKIRs, donors can be assigned either to KIR genotype A/A (ie, homozygous for A haplotypes) or to B/x (having 1 or 2 B haplotypes). The Cen and Tel regions can reassemble to form recombinant haplotypes. Genetic association studies suggest that an optimal alloreactive donor will be one with an iKIR-KIRL mismatch and who possesses multiple aKIRs (haplotype B) next to iKIRs. The KIR B content score mainly reflects the number of aKIRs, and the highest score is associated with the highest number of aKIRs. NK cells from such donors with a high KIR B content score should, in principle, exert strong alloreactive antileukemic NK cell activity, and it has been shown that transplantation with donors homozygous for Cen KIR B haplotypes is associated with the lowest level of relapse and highest overall survival.36,42,43  The KIR B content score can easily be determined by KIR genotyping. Note that the majority of haplotype B donors do not express all genes shown in the figure.

Close modal

There are two approaches to exploit the antileukemic effect of alloreactive NK cells. One is the adoptive transfer of NK cells from an alloreactive donor, with moderate lympho-depleting chemotherapy to induce homeostatic lymphocyte proliferation with transient expansion of the transferred allogeneic NK cells without the establishment of a permanent donor hematopoiesis. The other is the transplant of hematopoietic stem cells after myeloablative therapy and the permanent establishment of donor hematopoiesis and permanent engraftment of donor NK cells, thus establishing the donor KIR phenotype in the patient. The two approaches are depicted in Figure 4.

Figure 4

Two approaches for exploiting alloreactive NK cells. NK cells from a KIR-KIRL mismatched alloreactive donor or from umbilical cord blood (UCB) can be transferred directly or after ex vivo expansion after mild chemotherapy with transient NK cell proliferation. This will induce only a short-lived antileukemic effect. However, a patient can be permanently engrafted with hematopoietic stem cells from an alloreactive donor or UCB after myeloablative therapy, thus establishing a permanent donor KIR phenotype and antileukemic effect. TcRαβ, αβ T-cell receptor.

Figure 4

Two approaches for exploiting alloreactive NK cells. NK cells from a KIR-KIRL mismatched alloreactive donor or from umbilical cord blood (UCB) can be transferred directly or after ex vivo expansion after mild chemotherapy with transient NK cell proliferation. This will induce only a short-lived antileukemic effect. However, a patient can be permanently engrafted with hematopoietic stem cells from an alloreactive donor or UCB after myeloablative therapy, thus establishing a permanent donor KIR phenotype and antileukemic effect. TcRαβ, αβ T-cell receptor.

Close modal

The adoptive transfer of allogeneic NK cells in a nontransplant or transplant setting has been, and is currently being, investigated in clinical trials. In a pioneering clinical study, Miller and coworkers treated extensively lympho-depleted adult AML patients in the nontransplant setting with ex vivo expanded haploidentical NK cells and administered IL-2 in vivo to promote NK cell functionality.44  Remarkably, NK cells persisted and expanded in the subgroup of patients who had been treated with a high-intensity conditioning regimen and who exhibited high intrinsic IL-15 concentrations. In a pilot study involving 10 favorable-prognosis pediatric AML patients, Rubnitz and coworkers were able to show that the combination of low-dose immune suppression with the adoptive transfer of KIR-KIRL mismatched NK cells and in vivo IL-2 treatment after standard chemotherapy was safe and feasible, with long-term survival of all 10 participants.45  Our group recently demonstrated in 8 children with poor prognosis AML, BCP-ALL, and T-ALL that the infusion of ex vivo IL-15-stimulated CD3/CD19-depleted stem cell grafts (containing high numbers of NK cells) was safe and feasible in the haploidentical setting.46 

Because NK cells in the nontransplant setting are ultimately rejected, other phase 1 or 2 trials have used ex vivo expanded donor NK cells in patients after more-intensive conditioning. Expansion can be performed using either ex vivo cytokine stimulation or activation via the mbIL15-41BBL-expressing K562 transfectant.47  As sources, both peripheral blood mononuclear cells and umbilical cord blood cells have been used. Although the adoptive transfer of ex vivo expanded NK cells is, to date, considered safe, the clinical benefit has yet to be shown. Given that NK cells, early after transplant, are dysfunctional without the supportive application of IL-2,48  this cytokine was used in combination with adoptive NK cell transfer.44,45,49  However, to achieve in vivo NK cell expansion with IL-2 as the only cytokine, the necessary high doses of IL-2 may induce significant toxicity. Because low-dose IL-2 therapy significantly expands regulatory T-cell numbers and, as such, limits NK cell functionality, an alternative treatment strategy with IL-15 has been conceived. Considering that NK cells crucially depend on IL-15 to achieve optimal differentiation, survival, and effector function,50  the approach to administer IL-15 in vivo is particularly interesting for patients in whom NK cell maturation appears to be blocked in an immature state early after transplant. However, the first-in-human IL-15 study (in malignant melanoma and metastatic renal cell cancer patients) demonstrated significant IL-15 toxicities as a result of elevated proinflammatory cytokine levels,51  and the optimal doses have yet to be defined. Another cytokine-based strategy improves antitumor properties of NK cells using a brief in vitro priming with IL-12, IL-15, and IL-18.52  These so-called cytokine-activated memory-like NK cells are interesting insofar as they exhibit long-lasting increased antitumor properties. In this regard, upregulated IL-2 receptor (CD25) expression renders cytokine-activated memory-like NK cells particularly sensitive to picomolar concentrations of IL-2 and, through this, promotes enhanced IFN-γ production upon re-stimulation with cytokines or tumor cells.53  Interestingly, murine and preclinical in vitro studies suggest that NK cells are actively suppressed by regulatory T cells,54  and it has been assumed that this suppression also occurs early posttransplant. Hence, in a recent study by the Miller group, Bachanova et al55  used adoptive NK cell transfer and in vivo IL-2 administration but concomitantly depleted regulatory T cells with an IL-2 diphtheria-toxin fusion protein.

To predict whether a theoretically existing KIR-KIRL mismatch will translate into NK cell alloreactivity and, thus, increase GVL effects, it might be useful to integrate the complexity and the kinetics of NK cell reconstitution into the picture. Donor NK cells that reconstitute, develop, and mature in an HLA-disparate recipient will be shaped in the predominantly donor type–like hematopoietic niche56  and are, therefore, donor tolerant and recipient alloreactive, at least in the first months after transplant.26,33,40,57  Keeping the complex process of licensing or “education” of NK cells in mind, it is obvious that the density of donor-derived HLA class I molecules is crucial to the question of whether or not emerging NK cells will be alloreactive. In line with this, it has been postulated that the infusion of megadoses of stem cells will enable better acquisition of NK cell maturity than the infusion of usually applied doses.9  However, many aspects in the processes that are involved in the maturation and education of NK cell populations after HSCT are, at present, unclear. It has been shown that NK cells are among the first lymphocytes to reconstitute to desirable cell numbers (of more than 0.1 × 109/L CD56+ cells) within the first month after transplant, both in adults58,59  and in children,33,40,60  and that the reconstitution kinetics may inversely correlate to relapse probability.59,61  However, the phenotype and cytolytic activity of reconstituting NK cells significantly depends on the preconditioning regime, the source of the graft, and the mode of transplantation. NK cell reconstitution was shown to be impaired in patients who had been exposed to reduced-intensity (nonmyeloablative) conditioning regimens62  (but was enhanced in unrelated cord blood as compared with bone marrow recipients)63,64  and in patients who had received selectively CD3+ or αβ T-cell receptor T cell–depleted grafts that genuinely contain high numbers of donor-derived NK cells with a mature phenotypical and functional profile.33,65  However, despite the presence of acceptable numbers of NK cells, a large number of patients will exhibit a skewing of the NK cell repertoire to the extent that a phenotypically immature subset with little capacity for IFN-γ production (but with some cytotoxic function) will prevail.66  Of interest, individuals with a large pool of NKG2A+KIR NK cells, few cytotoxic CD3CD56dim NK cells, and little expression of iKIRs and NKp30 are prone to leukemic relapse.67,68  This would support the observation that KIR-negative unlicensed NK cells early after transplant are hyporesponsive (Figure 2D). In line with the definition that NK cell alloreactivity implies, the presence of iKIRs with varying HLA class I specificities, and the concomitant lack of CD94/NKG2A, analysis of the size and the characteristics of the iKIR NK cell subset is key to the question of which patients experience clinically relevant GVL effects.9,69  Although alloreactive NK cells have been shown to persist and expand in recipients for up to years,40  the size of alloreactive NK cell subsets varies somewhat unpredictably, both in donors and in patients. Phenotypically, the pool of alloreactive NK cells may, in the first place, comprise varying proportions KIR2DL1+, KIR2DL2/3+, KIR3DL1+, and/or KIR2DS1+ subsets. In this regard, KIR2DL1+ NK cells will be inhibited by HLA-C2 recipient’s cells, whereas they will lyse leukemia in C1/C1 recipients. KIR2DL2/3+ NK cells will recognize HLA-C1 but, peculiarly, also low-affinity HLA-C240 ; thus, leukemia of homozygous C2 recipients will only partially be lysed. KIR3DL1+ NK cells have HLA-A- and HLA-B-determined Bw4 supertypic specificity and will, therefore, lyse Bw4 leukemia. And lastly, KIR2DS1+ NK cells will kill leukemia in homozygous HLA-C2 recipients; however, only if the donor is HLA-C1 homozygous. Here, the activating signal of KIR2DS1 will overcome the inhibition of KIR2DL2/3, whereas in HLA-C2 donors, KIR2DS1 will dampen NK cell functionality to prevent autoreactivity.70  A summary of the KIR-KIRL combinations that effectively contribute to NK cell alloreactivity is shown in Table 1.

Table 1

KIR-KIRL combinations that effectively contribute to NK cell alloreactivity

KIR-KIRL interactionFunctional consequencesNK cell alloreactivity toward
KIR2DL1–HLA-C2 Inhibition in HLA-C2+ patients Lysis in HLA-C1+ patients 
KIR2DL2/3–HLA-C1 inhibition in HLA-C1+ patients Partial lysis in HLA-C2+ patients* 
KIR3DL1–HLA-Bw4 Inhibition in patients with Bw4+ supertypic specificity Lysis in Bw4 patients 
KIR2DS1–HLA-C2 Activation in HLA-C2+ patients Lysis in HLA-C2+ patients (only when donor is HLA-C1/C1) 
KIR-KIRL interactionFunctional consequencesNK cell alloreactivity toward
KIR2DL1–HLA-C2 Inhibition in HLA-C2+ patients Lysis in HLA-C1+ patients 
KIR2DL2/3–HLA-C1 inhibition in HLA-C1+ patients Partial lysis in HLA-C2+ patients* 
KIR3DL1–HLA-Bw4 Inhibition in patients with Bw4+ supertypic specificity Lysis in Bw4 patients 
KIR2DS1–HLA-C2 Activation in HLA-C2+ patients Lysis in HLA-C2+ patients (only when donor is HLA-C1/C1) 
*

See Pende et al.40 

We have observed that a substantial number of potentially alloreactive donor-derived NK cells expressing only a single iKIR were detectable after HLA-mismatched HSCT. Of interest, KIR2DL2/3 was predominantly expressed, irrespective of the patients’ HLA type. This may explain why patients homozygous for the C1 group (C1/C1) and, therefore, expressing more inhibitory ligands for KIR2DL2/3 had a poorer survival than patients with a C1/C2 or C2/C2 HLA type.33 

Next to these well-defined phenotypical properties of human NK cells with inherent potent GVL functionality, additional adaptive NK cell responses have been postulated when a link between posttransplant cytomegalovirus (CMV) infection and relapse protection was documented in AML patients.71,72  It was shown that CMV infection stably imprints the NK cell repertoire,73  thus promoting the clonal expansion of an NKG2C+aKIR+ NK cell compartment.57,74,75  This potentially alloreactive NK cell subset is CD56dim, simultaneously expresses CD57+, and secretes IFN-γ in response to K562 stimulation.76  At the molecular level, the methylation-induced silencing of FcεRγ, SYK, and EAT-2 DNA promoter regions triggers this adaptive CMV-induced conversion of NK cells.76  Functionally, CD56dimFcεRγ NK cells are prone to secrete cytokines and to exert antibody-dependent cellular cytotoxicity that is not restricted to CMV-infected target cells.76-78  It has been speculated that either MHC class I–like molecules (such as HLA-E whose expression is retained on leukemic cells67 ) trigger NK cell activity directly via NKG2C78  or virally encoded or so-far-unknown non-HLA ligands trigger other activating NK cell receptors71  (such as aKIRs,74  for example). Although the precise mechanisms of adaptive NK cell conversion remain elusive, a simplified view could be that in the context of viral infection, a proinflammatory stimulus disrupts the tumor-induced immunosuppression and promotes NK cell alloreactivity toward leukemia.

Despite many publications that provide evidence for the relevance of NK cell alloreactivity in the elimination of adult AML, but not BPC-ALL, less knowledge exists as to the significance of NK cell alloreactivity in the context of pediatric leukemia. In line with the data obtained in adult patients, pediatric AML appears to be a target of alloreactive NK cells.26,45,79,80  In contrast to adult BCP-ALL, pediatric BCP-ALL seems to be susceptible to NK cell–mediated target cell lysis.26,27,69,81  The difference in the susceptibility of adult and childhood BCP-ALL to NK cell–mediated lysis has been ascribed, in part, to differing expression of cell adhesion molecules of the β1 (CD29, CD49d) and β2 integrin (leukocyte functional antigen 1) family and the immunoglobulin superfamily (ICAM-1, leukocyte functional antigen 3),22,82  which essentially results in a reduced NK cell–target conjugate formation and activation in the case of adult BCP-ALL. However, with respect to KIR-KIRL incompatibility, it is probably more important that the surface density of MHC class I ligands is higher in pediatric BCP-ALL as compared with AML,79  although it has been demonstrated that the in vitro cytolytic activity of NK cells against BCP-ALL, in part, correlates to the extent of MHC expression.81,83  To allow the prediction of whether a given leukemia will indeed be the target of alloreactive NK cells, the preclinical testing should probably incorporate phenotypical and functional analyses on the clonal NK cell population level,26  as well as functional binding and redirected cytotoxicity assays in the presence of blocking antibodies to well-defined KIR epitopes,40,79  ideally using patient-specific leukemia as target cells. In this regard, the recent generation of antibodies that can discriminate KIR2DS1 from KIR2DL1, KIR3DS1 from KIR3DL1, and KIR2DS2 from KIR2DL3, but not KIR2DL2, has allowed a more precise analysis of potentially alloreactive NK cell subsets that a donor might express toward a given leukemia.84 

Why do alloreactive NK cells mediate GVL effects but, obviously, do not promote the induction of or even prevent GVHD12,85  in the context of ex vivo T cell–depleted HSCT? Experimental evidence obtained in mice demonstrates that alloreactive NK cells may lyse donor-derived antigen-presenting cells and T cells,12,22  may produce transforming growth factor β85  and, by this and other means, achieve control over the antigen-driven proliferation of CD4+86  and CD8+ T cells87  in the bone marrow compartment. Of interest, the targeted deletion of recipient dendritic cells or T cells critically depends on a process called trogocytosis, which enables the downstream acquisition of CCR7, a chemokine ligand which is required for the directed lymph node migration of licensed NK cells in response to CCL19 and CCL21.88  This event is negatively regulated by iKIRs and NKG2A but promoted by KIR2DS1.89  Because nonhematopoietic host tissues such as the skin, which is commonly affected by T cell–mediated GVHD, lack the expression of activating NK cell receptor ligands, the skin appears to be comparatively resistant to NK cell–mediated attack. It is currently assumed that, in some instances, NK cells might contribute to pathology after GVHD is established.90  In this regard, the mode of transplantation (ie, extent of T-cell depletion) and the mode of NK cell stimulation (ie, adoptive transfer with IL-15/4-1BBL activated and expanded NK cells) may affect GVHD occurrence.91 

One approach to counteract NK cell suppression induced by the KIR-KIRL interactions is the use of IPH2101, the first-in-class, nondepleting immunoglobulin G4 (IgG4) monoclonal antibody directed against KIR2DL1 and KIR2DL2/3. Indeed, IPH2101 therapy has been effectively able to restore NK cell alloreactivity in adult AML patients,92  and preliminary in vitro experiments also suggest effectiveness toward pediatric BCP-ALL specimens.69  An additional approach to activate alloreactive, but also nonalloreactive, NK cells is the use of monoclonal antibodies directed against antigens expressed on leukemic blasts. It has been shown that the activation of NK cells via the Fc-receptor CD16, which induces antibody-dependent cellular cytotoxicity, provides a strong activatory signal and overrides the inhibitory effects of the KIR-KIRL interaction.93  Because the CD19 antigen is widely expressed in BCP-ALL, an Fc-optimized anti-CD19 antibody was constructed and used posttransplant in selected high-risk patients with pediatric BCP-ALL.94  Other approaches to activate NK cells against BCP-ALL or AML in either the autologous or allogeneic setting, independent of their alloreactive status, might be the future clinical use of the CD16xCD1995  or CD16xCD33 bispecific killer engager96  or the trispecific killer engager CD16xCD19xCD22.97  NK cells and established NK cell lines such as NK-92 can also be redirected toward target antigens expressed on leukemic blasts by chimeric antigen receptors (CARs).98  It has been shown that CAR-NK cells can overcome inhibitory signals and induce specific killing of leukemic cells.99  Although the use of CAR-T cells has, so far, been restricted to autologous T cells, and although CAR-T cells might persist very long, resulting in on-target/off-tumor effects, CAR-NK cells could be adoptively and transiently transferred from allogeneic donors, or even off-the-shelf.100  The various approaches to overcome the KIR-KIRL-mediated inhibition and to increase the cytotoxicity toward leukemic cells in the autologous or allogeneic setting are shown in Figure 5.

Figure 5

Strategies to overcome the KIR-KIRL-mediated inhibition of NK cells and to increase their antileukemic response. (A) KIR-KIRL interaction is blocked by a monoclonal antibody, which abolishes the inhibitory signal. (B) KIR-KIRL-mediated inhibition is overridden by the activation of the Fc-receptor (CD16), with an antibody directed against an antigen expressed on leukemic blasts. (C-D) A bispecific killer engager and a trispecific killer engager activate NK cells via the Fc-receptor against antigens expressed on leukemia cells. (E) CAR-NK cells directed against the CD19 antigen are depicted.

Figure 5

Strategies to overcome the KIR-KIRL-mediated inhibition of NK cells and to increase their antileukemic response. (A) KIR-KIRL interaction is blocked by a monoclonal antibody, which abolishes the inhibitory signal. (B) KIR-KIRL-mediated inhibition is overridden by the activation of the Fc-receptor (CD16), with an antibody directed against an antigen expressed on leukemic blasts. (C-D) A bispecific killer engager and a trispecific killer engager activate NK cells via the Fc-receptor against antigens expressed on leukemia cells. (E) CAR-NK cells directed against the CD19 antigen are depicted.

Close modal

Substantial evidence exists that NK cell alloreactivity can be exploited in adult and pediatric patients with AML and probably also in children with BCP-ALL. However, the selection of a donor with a KIR-KIRL mismatch toward a given patient does not per se preclude the subsequent emergence of alloreactive NK cells with high GVL potential. In this regard, additionally expressed aKIRs, the mode of preconditioning, varying kinetics of NK cell reconstitution, prevailing cytokine levels, and presumably still undefined factors all contribute to NK cell alloreactivity. Therapeutic heightening of NK cell–mediated GVL effects can probably be achieved only with a multitiered approach that combines the optimal recognition of leukemia by KIR-KIRL mismatched NK cells, supportive cytokine or monoclonal antibody administration, and abrogation of NK cell–suppressive effects such as iKIR or regulatory T-cell signaling or transforming growth factor β–promoted cytokine signaling. A number of questions remain as to what the absolute number of NK cells is that is needed to achieve a clinical GVL response, how the different and sequentially appearing activating signals are integrated in the NK cell response to leukemia, what the direct functional evidence is for a role of multiple aKIRs in target cell recognition, and whether NK cells may adapt to leukemia in the sense of acquiring memory-like characteristics.

The authors thank Peter-Michael Weber for the graphic art.

This work was supported by the Deutsche Forschungsgemeinschaft grants KFO 183 and SFB 685 (M.C.A., P.L., and R.H.), the “Else Kröner-Fresenius-Stiftung” grant 2012 A296 (M.C.A. and R.H.), the Reinhold-Beitlich-Stiftung (P.L.), the “Stiftung für Krebskranke Kinder Tübingen e.V.,” the Jose-Carreras Leukemia Foundation, and the “Stefan-Morsch-Stiftung” (R.H.).

Contribution: M.C.A. and R.H. wrote the manuscript; and P.L. contributed data and critically reviewed and edited the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Rupert Handgretinger, Department of Pediatric Hematology and Oncology, University Children’s Hospital, Hoppe-Seyler-Str 1, D-72076 Tuebingen, Germany; e-mail: rupert.handgretinger@med.uni-tuebingen.de.

1
Spits
 
H
Artis
 
D
Colonna
 
M
, et al. 
Innate lymphoid cells--a proposal for uniform nomenclature.
Nat Rev Immunol
2013
, vol. 
13
 
2
(pg. 
145
-
149
)
2
Björkström
 
NK
Riese
 
P
Heuts
 
F
, et al. 
Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education.
Blood
2010
, vol. 
116
 
19
(pg. 
3853
-
3864
)
3
Kim
 
S
Poursine-Laurent
 
J
Truscott
 
SM
, et al. 
Licensing of natural killer cells by host major histocompatibility complex class I molecules.
Nature
2005
, vol. 
436
 
7051
(pg. 
709
-
713
)
4
Long
 
EO
Kim
 
HS
Liu
 
D
Peterson
 
ME
Rajagopalan
 
S
Controlling natural killer cell responses: integration of signals for activation and inhibition.
Annu Rev Immunol
2013
, vol. 
31
 (pg. 
227
-
258
)
5
Baer
 
MR
George
 
SL
Caligiuri
 
MA
, et al. 
Low-dose interleukin-2 immunotherapy does not improve outcome of patients age 60 years and older with acute myeloid leukemia in first complete remission: Cancer and Leukemia Group B Study 9720.
J Clin Oncol
2008
, vol. 
26
 
30
(pg. 
4934
-
4939
)
6
Burns
 
LJ
Weisdorf
 
DJ
DeFor
 
TE
, et al. 
IL-2-based immunotherapy after autologous transplantation for lymphoma and breast cancer induces immune activation and cytokine release: a phase I/II trial.
Bone Marrow Transplant
2003
, vol. 
32
 
2
(pg. 
177
-
186
)
7
Moretta
 
A
Bottino
 
C
Pende
 
D
, et al. 
Identification of four subsets of human CD3-CD16+ natural killer (NK) cells by the expression of clonally distributed functional surface molecules: correlation between subset assignment of NK clones and ability to mediate specific alloantigen recognition.
J Exp Med
1990
, vol. 
172
 
6
(pg. 
1589
-
1598
)
8
Parham
 
P
MHC class I molecules and KIRs in human history, health and survival.
Nat Rev Immunol
2005
, vol. 
5
 
3
(pg. 
201
-
214
)
9
Moretta
 
L
Locatelli
 
F
Pende
 
D
Marcenaro
 
E
Mingari
 
MC
Moretta
 
A
Killer Ig-like receptor-mediated control of natural killer cell alloreactivity in haploidentical hematopoietic stem cell transplantation.
Blood
2011
, vol. 
117
 
3
(pg. 
764
-
771
)
10
Hsu
 
KC
Liu
 
XR
Selvakumar
 
A
Mickelson
 
E
O’Reilly
 
RJ
Dupont
 
B
Killer Ig-like receptor haplotype analysis by gene content: evidence for genomic diversity with a minimum of six basic framework haplotypes, each with multiple subsets.
J Immunol
2002
, vol. 
169
 
9
(pg. 
5118
-
5129
)
11
Vilches
 
C
Parham
 
P
KIR: diverse, rapidly evolving receptors of innate and adaptive immunity.
Annu Rev Immunol
2002
, vol. 
20
 (pg. 
217
-
251
)
12
Ruggeri
 
L
Capanni
 
M
Urbani
 
E
, et al. 
Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
Science
2002
, vol. 
295
 
5562
(pg. 
2097
-
2100
)
13
Ruggeri
 
L
Mancusi
 
A
Capanni
 
M
, et al. 
Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value.
Blood
2007
, vol. 
110
 
1
(pg. 
433
-
440
)
14
Giebel
 
S
Locatelli
 
F
Lamparelli
 
T
, et al. 
Survival advantage with KIR ligand incompatibility in hematopoietic stem cell transplantation from unrelated donors.
Blood
2003
, vol. 
102
 
3
(pg. 
814
-
819
)
15
Willemze
 
R
Rodrigues
 
CA
Labopin
 
M
, et al. 
Eurocord-Netcord and Acute Leukaemia Working Party of the EBMT
KIR-ligand incompatibility in the graft-versus-host direction improves outcomes after umbilical cord blood transplantation for acute leukemia.
Leukemia
2009
, vol. 
23
 
3
(pg. 
492
-
500
)
16
Beelen
 
DW
Ottinger
 
HD
Ferencik
 
S
, et al. 
Genotypic inhibitory killer immunoglobulin-like receptor ligand incompatibility enhances the long-term antileukemic effect of unmodified allogeneic hematopoietic stem cell transplantation in patients with myeloid leukemias.
Blood
2005
, vol. 
105
 
6
(pg. 
2594
-
2600
)
17
Davies
 
SM
Ruggieri
 
L
DeFor
 
T
, et al. 
Evaluation of KIR ligand incompatibility in mismatched unrelated donor hematopoietic transplants.
Blood
2002
, vol. 
100
 
10
(pg. 
3825
-
3827
)
18
Farag
 
SS
Bacigalupo
 
A
Eapen
 
M
, et al. 
KIR Study Group, Center for International Blood and Marrow Transplantation Research
The effect of KIR ligand incompatibility on the outcome of unrelated donor transplantation: a report from the center for international blood and marrow transplant research, the European blood and marrow transplant registry, and the Dutch registry.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
8
(pg. 
876
-
884
)
19
Lowe
 
EJ
Turner
 
V
Handgretinger
 
R
, et al. 
T-cell alloreactivity dominates natural killer cell alloreactivity in minimally T-cell-depleted HLA-non-identical paediatric bone marrow transplantation.
Br J Haematol
2003
, vol. 
123
 
2
(pg. 
323
-
326
)
20
Brunstein
 
CG
Wagner
 
JE
Weisdorf
 
DJ
, et al. 
Negative effect of KIR alloreactivity in recipients of umbilical cord blood transplant depends on transplantation conditioning intensity.
Blood
2009
, vol. 
113
 
22
(pg. 
5628
-
5634
)
21
Weisdorf
 
D
Cooley
 
S
Devine
 
S
, et al. 
T cell-depleted partial matched unrelated donor transplant for advanced myeloid malignancy: KIR ligand mismatch and outcome.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
6
(pg. 
937
-
943
)
22
Ruggeri
 
L
Capanni
 
M
Casucci
 
M
, et al. 
Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation.
Blood
1999
, vol. 
94
 
1
(pg. 
333
-
339
)
23
Bishara
 
A
De Santis
 
D
Witt
 
CC
, et al. 
The beneficial role of inhibitory KIR genes of HLA class I NK epitopes in haploidentically mismatched stem cell allografts may be masked by residual donor-alloreactive T cells causing GVHD.
Tissue Antigens
2004
, vol. 
63
 
3
(pg. 
204
-
211
)
24
Huang
 
XJ
Zhao
 
XY
Liu
 
DH
Liu
 
KY
Xu
 
LP
Deleterious effects of KIR ligand incompatibility on clinical outcomes in haploidentical hematopoietic stem cell transplantation without in vitro T-cell depletion.
Leukemia
2007
, vol. 
21
 
4
(pg. 
848
-
851
)
25
Sobecks
 
RM
Wang
 
T
Askar
 
M
, et al. 
Impact of KIR and HLA genotypes on outcomes after reduced-intensity conditioning hematopoietic cell transplantation.
Biol Blood Marrow Transplant
2015
, vol. 
21
 
9
(pg. 
1589
-
1596
)
26
Leung
 
W
Iyengar
 
R
Turner
 
V
, et al. 
Determinants of antileukemia effects of allogeneic NK cells.
J Immunol
2004
, vol. 
172
 
1
(pg. 
644
-
650
)
27
Leung
 
W
Iyengar
 
R
Triplett
 
B
, et al. 
Comparison of killer Ig-like receptor genotyping and phenotyping for selection of allogeneic blood stem cell donors.
J Immunol
2005
, vol. 
174
 
10
(pg. 
6540
-
6545
)
28
Hsu
 
KC
Keever-Taylor
 
CA
Wilton
 
A
, et al. 
Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes.
Blood
2005
, vol. 
105
 
12
(pg. 
4878
-
4884
)
29
Hsu
 
KC
Gooley
 
T
Malkki
 
M
, et al. 
International Histocompatibility Working Group
KIR ligands and prediction of relapse after unrelated donor hematopoietic cell transplantation for hematologic malignancy.
Biol Blood Marrow Transplant
2006
, vol. 
12
 
8
(pg. 
828
-
836
)
30
Arima
 
N
Nakamura
 
F
Yabe
 
T
, et al. 
 
Influence of differently licensed KIR2DL1-positive natural killer cells in transplant recipients with acute leukemia: a Japanese National Registry study. Biol Blood Marrow Transplant. 2016;22(3):423-431
31
Gagne
 
K
Brizard
 
G
Gueglio
 
B
, et al. 
Relevance of KIR gene polymorphisms in bone marrow transplantation outcome.
Hum Immunol
2002
, vol. 
63
 
4
(pg. 
271
-
280
)
32
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
, vol. 
16
 
4
(pg. 
533
-
542
)
33
Pfeiffer
 
MM
Feuchtinger
 
T
Teltschik
 
HM
, et al. 
Reconstitution of natural killer cell receptors influences natural killer activity and relapse rate after haploidentical transplantation of T- and B-cell depleted grafts in children.
Haematologica
2010
, vol. 
95
 
8
(pg. 
1381
-
1388
)
34
Lang
 
P
Teltschik
 
HM
Feuchtinger
 
T
, et al. 
Transplantation of CD3/CD19 depleted allografts from haploidentical family donors in paediatric leukaemia.
Br J Haematol
2014
, vol. 
165
 
5
(pg. 
688
-
698
)
35
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
, vol. 
179
 
2
(pg. 
854
-
868
)
36
Cooley
 
S
Trachtenberg
 
E
Bergemann
 
TL
, et al. 
Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia.
Blood
2009
, vol. 
113
 
3
(pg. 
726
-
732
)
37
Cooley
 
S
Weisdorf
 
DJ
Guethlein
 
LA
, et al. 
Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia.
Blood
2010
, vol. 
116
 
14
(pg. 
2411
-
2419
)
38
Venstrom
 
JM
Pittari
 
G
Gooley
 
TA
, et al. 
HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1.
N Engl J Med
2012
, vol. 
367
 
9
(pg. 
805
-
816
)
39
Mancusi
 
A
Ruggeri
 
L
Urbani
 
E
, et al. 
Haploidentical hematopoietic transplantation from KIR ligand-mismatched donors with activating KIRs reduces nonrelapse mortality.
Blood
2015
, vol. 
125
 
20
(pg. 
3173
-
3182
)
40
Pende
 
D
Marcenaro
 
S
Falco
 
M
, et al. 
Anti-leukemia activity of alloreactive NK cells in KIR ligand-mismatched haploidentical HSCT for pediatric patients: evaluation of the functional role of activating KIR and redefinition of inhibitory KIR specificity.
Blood
2009
, vol. 
113
 
13
(pg. 
3119
-
3129
)
41
Triplett
 
BM
Horwitz
 
EM
Iyengar
 
R
, et al. 
Effects of activating NK cell receptor expression and NK cell reconstitution on the outcomes of unrelated donor hematopoietic cell transplantation for hematologic malignancies.
Leukemia
2009
, vol. 
23
 
7
(pg. 
1278
-
1287
)
42
Cooley
 
S
Weisdorf
 
DJ
Guethlein
 
LA
, et al. 
Donor killer cell Ig-like receptor B haplotypes, recipient HLA-C1, and HLA-C mismatch enhance the clinical benefit of unrelated transplantation for acute myelogenous leukemia.
J Immunol
2014
, vol. 
192
 
10
(pg. 
4592
-
4600
)
43
Oevermann
 
L
Michaelis
 
SU
Mezger
 
M
, et al. 
KIR B haplotype donors confer a reduced risk for relapse after haploidentical transplantation in children with ALL.
Blood
2014
, vol. 
124
 
17
(pg. 
2744
-
2747
)
44
Miller
 
JS
Soignier
 
Y
Panoskaltsis-Mortari
 
A
, et al. 
Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer.
Blood
2005
, vol. 
105
 
8
(pg. 
3051
-
3057
)
45
Rubnitz
 
JE
Inaba
 
H
Ribeiro
 
RC
, et al. 
NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia.
J Clin Oncol
2010
, vol. 
28
 
6
(pg. 
955
-
959
)
46
Pfeiffer
 
MM
Schumm
 
M
Müller
 
I
Handgretinger
 
R
Lang
 
P
IL-15-stimulated CD3/CD19-depleted stem-cell boosts in relapsed pediatric patients after haploidentical SCT.
Leukemia
2012
, vol. 
26
 
11
(pg. 
2435
-
2439
)
47
Fujisaki
 
H
Kakuda
 
H
Shimasaki
 
N
, et al. 
Expansion of highly cytotoxic human natural killer cells for cancer cell therapy.
Cancer Res
2009
, vol. 
69
 
9
(pg. 
4010
-
4017
)
48
Pittari
 
G
Fregni
 
G
Roguet
 
L
, et al. 
Early evaluation of natural killer activity in post-transplant acute myeloid leukemia patients.
Bone Marrow Transplant
2010
, vol. 
45
 
5
(pg. 
862
-
871
)
49
Curti
 
A
Ruggeri
 
L
D’Addio
 
A
, et al. 
Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients.
Blood
2011
, vol. 
118
 
12
(pg. 
3273
-
3279
)
50
Huntington
 
ND
The unconventional expression of IL-15 and its role in NK cell homeostasis.
Immunol Cell Biol
2014
, vol. 
92
 
3
(pg. 
210
-
213
)
51
Conlon
 
KC
Lugli
 
E
Welles
 
HC
, et al. 
Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer.
J Clin Oncol
2015
, vol. 
33
 
1
(pg. 
74
-
82
)
52
Romee
 
R
Schneider
 
SE
Leong
 
JW
, et al. 
Cytokine activation induces human memory-like NK cells.
Blood
2012
, vol. 
120
 
24
(pg. 
4751
-
4760
)
53
Leong
 
JW
Chase
 
JM
Romee
 
R
, et al. 
Preactivation with IL-12, IL-15, and IL-18 induces CD25 and a functional high-affinity IL-2 receptor on human cytokine-induced memory-like natural killer cells.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
4
(pg. 
463
-
473
)
54
Ghiringhelli
 
F
Ménard
 
C
Terme
 
M
, et al. 
CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner.
J Exp Med
2005
, vol. 
202
 
8
(pg. 
1075
-
1085
)
55
Bachanova
 
V
Cooley
 
S
Defor
 
TE
, et al. 
Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein.
Blood
2014
, vol. 
123
 
25
(pg. 
3855
-
3863
)
56
Sykes
 
M
Harty
 
MW
Karlhofer
 
FM
Pearson
 
DA
Szot
 
G
Yokoyama
 
W
Hematopoietic cells and radioresistant host elements influence natural killer cell differentiation.
J Exp Med
1993
, vol. 
178
 
1
(pg. 
223
-
229
)
57
Della Chiesa
 
M
Falco
 
M
Bertaina
 
A
, et al. 
Human cytomegalovirus infection promotes rapid maturation of NK cells expressing activating killer Ig-like receptor in patients transplanted with NKG2C-/- umbilical cord blood.
J Immunol
2014
, vol. 
192
 
4
(pg. 
1471
-
1479
)
58
Servais
 
S
Lengline
 
E
Porcher
 
R
, et al. 
Long-term immune reconstitution and infection burden after mismatched hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
2014
, vol. 
20
 
4
(pg. 
507
-
517
)
59
Klyuchnikov
 
E
Asenova
 
S
Kern
 
W
, et al. 
Post-transplant immune reconstitution after unrelated allogeneic stem cell transplant in patients with acute myeloid leukemia.
Leuk Lymphoma
2010
, vol. 
51
 
8
(pg. 
1450
-
1463
)
60
Rénard
 
C
Barlogis
 
V
Mialou
 
V
, et al. 
Lymphocyte subset reconstitution after unrelated cord blood or bone marrow transplantation in children.
Br J Haematol
2011
, vol. 
152
 
3
(pg. 
322
-
330
)
61
Lang
 
P
Pfeiffer
 
M
Teltschik
 
HM
, et al. 
Natural killer cell activity influences outcome after T cell depleted stem cell transplantation from matched unrelated and haploidentical donors.
Best Pract Res Clin Haematol
2011
, vol. 
24
 
3
(pg. 
403
-
411
)
62
Pical-Izard
 
C
Crocchiolo
 
R
Granjeaud
 
S
, et al. 
Reconstitution of natural killer cells in HLA-matched HSCT after reduced-intensity conditioning: impact on clinical outcome.
Biol Blood Marrow Transplant
2015
, vol. 
21
 
3
(pg. 
429
-
439
)
63
Bartelink
 
IH
Belitser
 
SV
Knibbe
 
CA
, et al. 
Immune reconstitution kinetics as an early predictor for mortality using various hematopoietic stem cell sources in children.
Biol Blood Marrow Transplant
2013
, vol. 
19
 
2
(pg. 
305
-
313
)
64
Charrier
 
E
Cordeiro
 
P
Brito
 
RM
, et al. 
Reconstitution of maturating and regulatory lymphocyte subsets after cord blood and BMT in children.
Bone Marrow Transplant
2013
, vol. 
48
 
3
(pg. 
376
-
382
)
65
Lang
 
P
Feuchtinger
 
T
Teltschik
 
HM
, et al. 
Improved immune recovery after transplantation of TCRαβ/CD19-depleted allografts from haploidentical donors in pediatric patients.
Bone Marrow Transplant
2015
, vol. 
50
 
Suppl 2
(pg. 
S6
-
S10
)
66
Foley
 
B
Cooley
 
S
Verneris
 
MR
, et al. 
NK cell education after allogeneic transplantation: dissociation between recovery of cytokine-producing and cytotoxic functions.
Blood
2011
, vol. 
118
 
10
(pg. 
2784
-
2792
)
67
Nguyen
 
S
Dhedin
 
N
Vernant
 
JP
, et al. 
NK-cell reconstitution after haploidentical hematopoietic stem-cell transplantations: immaturity of NK cells and inhibitory effect of NKG2A override GvL effect.
Blood
2005
, vol. 
105
 
10
(pg. 
4135
-
4142
)
68
Vago
 
L
Forno
 
B
Sormani
 
MP
, et al. 
Temporal, quantitative, and functional characteristics of single-KIR-positive alloreactive natural killer cell recovery account for impaired graft-versus-leukemia activity after haploidentical hematopoietic stem cell transplantation.
Blood
2008
, vol. 
112
 
8
(pg. 
3488
-
3499
)
69
Kübler
 
A
Woiterski
 
J
Witte
 
KE
, et al. 
Both mature KIR+ and immature KIR- NK cells control pediatric acute B-cell precursor leukemia in NOD.Cg-Prkdcscid IL2rgtmWjl/Sz mice.
Blood
2014
, vol. 
124
 
26
(pg. 
3914
-
3923
)
70
Fauriat
 
C
Ivarsson
 
MA
Ljunggren
 
HG
Malmberg
 
KJ
Michaëlsson
 
J
Education of human natural killer cells by activating killer cell immunoglobulin-like receptors.
Blood
2010
, vol. 
115
 
6
(pg. 
1166
-
1174
)
71
Elmaagacli
 
AH
Steckel
 
NK
Koldehoff
 
M
, et al. 
Early human cytomegalovirus replication after transplantation is associated with a decreased relapse risk: evidence for a putative virus-versus-leukemia effect in acute myeloid leukemia patients.
Blood
2011
, vol. 
118
 
5
(pg. 
1402
-
1412
)
72
Green
 
ML
Leisenring
 
WM
Xie
 
H
, et al. 
CMV reactivation after allogeneic HCT and relapse risk: evidence for early protection in acute myeloid leukemia.
Blood
2013
, vol. 
122
 
7
(pg. 
1316
-
1324
)
73
Gumá
 
M
Angulo
 
A
Vilches
 
C
Gómez-Lozano
 
N
Malats
 
N
López-Botet
 
M
Imprint of human cytomegalovirus infection on the NK cell receptor repertoire.
Blood
2004
, vol. 
104
 
12
(pg. 
3664
-
3671
)
74
Béziat
 
V
Liu
 
LL
Malmberg
 
JA
, et al. 
NK cell responses to cytomegalovirus infection lead to stable imprints in the human KIR repertoire and involve activating KIRs.
Blood
2013
, vol. 
121
 
14
(pg. 
2678
-
2688
)
75
Cichocki
 
F
Cooley
 
S
Davis
 
Z
, et al. 
CD56(dim)CD57(+)NKG2C(+) NK cell expansion is associated with reduced leukemia relapse after reduced intensity HCT.
Leukemia
2016
, vol. 
30
 
2
(pg. 
456
-
463
)
76
Schlums
 
H
Cichocki
 
F
Tesi
 
B
, et al. 
Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function.
Immunity
2015
, vol. 
42
 
3
(pg. 
443
-
456
)
77
Zhang
 
T
Scott
 
JM
Hwang
 
I
Kim
 
S
Cutting edge: antibody-dependent memory-like NK cells distinguished by FcRγ deficiency.
J Immunol
2013
, vol. 
190
 
4
(pg. 
1402
-
1406
)
78
Foley
 
B
Cooley
 
S
Verneris
 
MR
, et al. 
Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function.
Blood
2012
, vol. 
119
 
11
(pg. 
2665
-
2674
)
79
Pende
 
D
Spaggiari
 
GM
Marcenaro
 
S
, et al. 
Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112).
Blood
2005
, vol. 
105
 
5
(pg. 
2066
-
2073
)
80
Schlegel
 
P
Ditthard
 
K
Lang
 
P
, et al. 
 
NKG2D signaling leads to NK cell mediated lysis of childhood AML. J Immunol Res. 2015;2015:473175
81
Pfeiffer
 
M
Schumm
 
M
Feuchtinger
 
T
Dietz
 
K
Handgretinger
 
R
Lang
 
P
Intensity of HLA class I expression and KIR-mismatch determine NK-cell mediated lysis of leukaemic blasts from children with acute lymphatic leukaemia.
Br J Haematol
2007
, vol. 
138
 
1
(pg. 
97
-
100
)
82
Mengarelli
 
A
Zarcone
 
D
Caruso
 
R
, et al. 
Adhesion molecule expression, clinical features and therapy outcome in childhood acute lymphoblastic leukemia.
Leuk Lymphoma
2001
, vol. 
40
 
5-6
(pg. 
625
-
630
)
83
Feuchtinger
 
T
Pfeiffer
 
M
Pfaffle
 
A
, et al. 
Cytolytic activity of NK cell clones against acute childhood precursor-B-cell leukaemia is influenced by HLA class I expression on blasts and the differential KIR phenotype of NK clones.
Bone Marrow Transplant
2009
, vol. 
43
 
11
(pg. 
875
-
881
)
84
Vitale
 
M
Carlomagno
 
S
Falco
 
M
, et al. 
Isolation of a novel KIR2DL3-specific mAb: comparative analysis of the surface distribution and function of KIR2DL2, KIR2DL3 and KIR2DS2.
Int Immunol
2004
, vol. 
16
 
10
(pg. 
1459
-
1466
)
85
Asai
 
O
Longo
 
DL
Tian
 
ZG
, et al. 
Suppression of graft-versus-host disease and amplification of graft-versus-tumor effects by activated natural killer cells after allogeneic bone marrow transplantation.
J Clin Invest
1998
, vol. 
101
 
9
(pg. 
1835
-
1842
)
86
Noval Rivas
 
M
Hazzan
 
M
Weatherly
 
K
Gaudray
 
F
Salmon
 
I
Braun
 
MY
NK cell regulation of CD4 T cell-mediated graft-versus-host disease.
J Immunol
2010
, vol. 
184
 
12
(pg. 
6790
-
6798
)
87
Zecher
 
D
Li
 
Q
Oberbarnscheidt
 
MH
, et al. 
NK cells delay allograft rejection in lymphopenic hosts by downregulating the homeostatic proliferation of CD8+ T cells.
J Immunol
2010
, vol. 
184
 
12
(pg. 
6649
-
6657
)
88
Marcenaro
 
E
Cantoni
 
C
Pesce
 
S
, et al. 
Uptake of CCR7 and acquisition of migratory properties by human KIR+ NK cells interacting with monocyte-derived DC or EBV cell lines: regulation by KIR/HLA-class I interaction.
Blood
2009
, vol. 
114
 
19
(pg. 
4108
-
4116
)
89
Marcenaro
 
E
Pesce
 
S
Sivori
 
S
Carlomagno
 
S
Moretta
 
L
Moretta
 
A
KIR2DS1-dependent acquisition of CCR7 and migratory properties by human NK cells interacting with allogeneic HLA-C2+ DCs or T-cell blasts.
Blood
2013
, vol. 
121
 
17
(pg. 
3396
-
3401
)
90
Foley
 
B
Felices
 
M
Cichocki
 
F
Cooley
 
S
Verneris
 
MR
Miller
 
JS
The biology of NK cells and their receptors affects clinical outcomes after hematopoietic cell transplantation (HCT).
Immunol Rev
2014
, vol. 
258
 
1
(pg. 
45
-
63
)
91
Shah
 
NN
Baird
 
K
Delbrook
 
CP
, et al. 
Acute GVHD in patients receiving IL-15/4-1BBL activated NK cells following T-cell-depleted stem cell transplantation.
Blood
2015
, vol. 
125
 
5
(pg. 
784
-
792
)
92
Vey
 
N
Bourhis
 
JH
Boissel
 
N
, et al. 
A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission.
Blood
2012
, vol. 
120
 
22
(pg. 
4317
-
4323
)
93
Lang
 
P
Barbin
 
K
Feuchtinger
 
T
, et al. 
Chimeric CD19 antibody mediates cytotoxic activity against leukemic blasts with effector cells from pediatric patients who received T-cell-depleted allografts.
Blood
2004
, vol. 
103
 
10
(pg. 
3982
-
3985
)
94
Seidel
 
UJ
Vogt
 
F
Grosse-Hovest
 
L
Jung
 
G
Handgretinger
 
R
Lang
 
P
γδ T cell-mediated antibody-dependent cellular cytotoxicity with CD19 antibodies assessed by an impedance-based label-free real-time cytotoxicity assay.
Front Immunol
2014
, vol. 
5
 pg. 
618
 
95
Kellner
 
C
Bruenke
 
J
Horner
 
H
, et al. 
Heterodimeric bispecific antibody-derivatives against CD19 and CD16 induce effective antibody-dependent cellular cytotoxicity against B-lymphoid tumor cells.
Cancer Lett
2011
, vol. 
303
 
2
(pg. 
128
-
139
)
96
Gleason
 
MK
Ross
 
JA
Warlick
 
ED
, et al. 
CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets.
Blood
2014
, vol. 
123
 
19
(pg. 
3016
-
3026
)
97
Gleason
 
MK
Verneris
 
MR
Todhunter
 
DA
, et al. 
Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production.
Mol Cancer Ther
2012
, vol. 
11
 
12
(pg. 
2674
-
2684
)
98
Glienke
 
W
Esser
 
R
Priesner
 
C
, et al. 
Advantages and applications of CAR-expressing natural killer cells.
Front Pharmacol
2015
, vol. 
6
 pg. 
21
 
99
Imai
 
C
Iwamoto
 
S
Campana
 
D
Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells.
Blood
2005
, vol. 
106
 
1
(pg. 
376
-
383
)
100
Hermanson
 
DL
Kaufman
 
DS
Utilizing chimeric antigen receptors to direct natural killer cell activity.
Front Immunol
2015
, vol. 
6
 pg. 
195
 
Sign in via your Institution