Open access peer-reviewed chapter

Clinical Applications of Natural Killer Cells

Written By

Yui Harada, Koji Teraishi, Minori Ishii, Hiroshi Ban and Yoshikazu Yonemitsu

Submitted: 02 December 2016 Reviewed: 05 April 2017 Published: 13 December 2017

DOI: 10.5772/intechopen.68991

From the Edited Volume

Natural Killer Cells

Edited by Mourad Aribi

Chapter metrics overview

2,412 Chapter Downloads

View Full Metrics

Abstract

Natural killer (NK) cells are an essential component of the innate immune system, and they play a crucial role in immunity against malignancies. Recent advances in our understanding of NK cell biology have paved the way for new therapeutic strategies based on NK cells for the treatment of various cancers. In this section, we will focus on NK cell immunotherapy, including the enhancement of antibody‐dependent cellular cytotoxicity, the manipulation of receptor‐mediated activation, inclusion criteria based on killer cell immunoglobulin‐like receptor (KIR) ligand mismatches, and adoptive immunotherapy with ex vivo expanded chimeric antigen receptor (CAR)‐engineered or engager‐modified NK cells. In contrast to T lymphocytes, donor NK cells do not attack any recipient tissues based on allogeneic human leukocyte antigens (HLAs), suggesting that NK‐mediated antitumor effects may be achieved without the risk of graft‐versus‐host disease (GvHD). Despite reports of clinical efficacy, the application of NK cell immunotherapy is limited. Developing strategies for manipulating NK cell products, host factors, and tumor targets are thus current subjects of diligent study. Research into the biology of NK cells has indicated that NK cell immunotherapy has the potential to become the forefront of cancer immunotherapy in the coming years.

Keywords

  • NK cell
  • KIRs
  • immunotherapy
  • HSC transplantation
  • genetic modification

1. Introduction

Natural killer (NK) cells have been used in clinical studies in order to treat various malignancies. Missing‐self is one of the mechanisms of the NK cell response that works by the detection of the loss of autologous major histocompatibility complex (MHC) class I expression.

We will mention four elements of NK cells in this chapter: (1) mechanisms of NK cells; (2) activation of NK cells; (3) inclusion criteria based on KIR ligand mismatches; and (4) genetic modifications ex vivo.

NK cells, which are thought to have emerged much later than B cells and T cells based on the evolutionary convergence of variable receptors, rarely cause autoimmune diseases. We will introduce the applicability of the NK cell to cancer treatment.

2. Activation of NK cells

NK cell‐based immunotherapy has been explored for decades. Several animal experiments indicated the potential efficacy of NK cells in cancer treatment [13]. However, ex vivo NK cell expansion techniques have been insufficient regarding the numbers of cells, purity and antitumor activity to use in clinical settings. NK cells comprise only a minor population (i.e., 5–15% of peripheral lymphocytes), and only a small number of NK cells is isolated after a typical apheresis procedure. For example, approx. 77.8 ± 14.4 × 109/L (range 62.7–95.9 × 109/L) of leukapheretic products are the result of a single apheresis of peripheral blood in a normal adult human with a mean percentage of lymphocytes of 59.8% ± 6.1 (range 53.9–66.4%), and subsequently 5–10 × 108 NK cells can be obtained [4]. On the other hand, at least 2 × 107/kg or 6 × 106–6.5 × 109/body NK cells are required for each effective injection with multiple administrations [5, 6]. In addition, the reported engraftment period of NK cells was 2–189 days (median 10 days), which showed no correlation with the number of NK cells administered [7]. The NK cell infusion should thus be repeated in order to maintain a sufficient number of NK cells meeting clinical requirements, but this would be a burden on patients.

Scientists have been working to develop various methods for proliferating NK cells ex vivo with high cytotoxicity and high purity. Several studies used an anti‐CD3 antibody (clone: OKT3) in the first few days of culture for the activation of autologous T cells to help the NK cell expansion, subsequently producing high numbers of undesirable T cells or NKT cells in the final product [810]. However, particularly in haploidentical NK cell transplantation, T cells must be excluded prior to the infusion in order to prevent graft versus host disease (GvHD). Other studies removed CD3+ cells by magnetic beads with or without CD56‐positive selection at the beginning of culture [1113].

To acquire highly purified and expanded NK cells, an initial efficient depletion (<1%) of CD3+ cells and a relatively long‐term culture (over 12 days) seem to be essential [12, 13]. Because only a minor fraction of circulating NK cells is reactive to target cells (tumor cells) in vitro, primary NK cells show insufficient cytotoxicity [14]. Various types of stimulation have thus been reported to enable NK cells to achieve their full effector potential, such as interleukin (IL)‐15 produced by dendritic cells (DCs) [15] or macrophages [16], IL‐2 [17], IL‐12 [18], IL‐18 [19] and IL‐21 [20]. Currently, the additional cytokines used in the cultivation of NK cells include IL‐2, IL‐15, and IL‐21. IL‐2, IL‐15, and IL‐21 share the receptor subunits IL‐2/15Rβ and common γ chain [21] on the NK cell surface and have a synergetic effect. Use of IL‐2 combined with IL‐15 for cultivation leads to good viability and good proliferation of NK cells [22]. IL‐2 is also important for NK cell infiltration and killing, and IL‐15 is important for both NK cell maturation and survival [21]. IL‐2 and IL‐15 induce the expression of KIRs and activating receptors (NKG2D and NKp44) on NK cell surface [23]. IL‐21 modifies the expression of killer cell immunoglobulin‐like receptors (KIRs) and NKp44 by reducing expression of DAP‐12, subsequently promote cell maturation, the ability of killing and survival [22, 23]. Several experiments used feeder cells to provide essential stimulation for NK cell cultivation through cytokine production or cell‐to‐cell contact [20, 23, 24]. The various cytokines and feeder cells used in some clinical trials are mentioned in a later section of this article.

2.1. Killer cell immunoglobulin‐like receptors (KIRs)

NK cells express KIRs, most of which are inhibitory (partially activating) receptors that recognized MHC class I molecules. In the 1980s, KIRs were first described as explaining the NK cell‐mediated rejection of allogeneic bone marrow transplants from a homozygous donor to a hemizygous host in lymphoma and in F1‐hybrid anti‐parental resistance in a rodent model [25]. In these situations, the graft fails to express at least one MHC class I allele of the host, and NK cells are highly capable of identifying the difference, thus causing rejection. This can be explained by the concept that NK cells have inhibitory receptors and begin to attack target cells if they do not express ligands that interact with the specific inhibitory receptors. This phenomenon was termed the “missing‐self hypothesis”, which is now accepted as one of the complex target recognition mechanisms of NK cells.

2.1.1. Regulation of NK cell activity

The activation of NK cells is regulated by various receptors including KIRs, CD94–NKG2 family, leukocyte immunoglobulin‐like receptors (LILRs), natural cytotoxicity receptors (NCRs), and FcγRIIIa (CD16) [2628]. Among these receptors, KIRs, CD94/NKG2A heterodimers, and LILRs belong to the large family of inhibitory receptors of MHC class I, mediating NK cell function by signaling through intracytoplasmic immunoreceptor tyrosine‐based inhibition motifs (ITIMs) [29]. Each NK cell has a threshold of activation through a balance of total stimulation between inhibitory and activating signals [30] (Figure 1). In other words, NK cells selectively kill target cells that down‐regulate MHC class I molecules and/or up‐regulate other activating ligands [28] such as MHC class I chain (MIC)‐related antigens MICA, MICB and UL‐16 binding protein (ULBP). MICA, MICB, ULBPs are ligands of NKG2D homodimer, which belong to C‐type lectin receptor NKG2 family expressed on the surface of NK cells and CD8+ T‐lymphocytes. NKG2A/CD94 and NKG2B/CD94 heterodimers transmit inhibitory signals, while NKG2C/CD94, NKG2E/CD94, NKG2H/CD94 heterodimers and NKG2D homodimer are activating receptors.

Figure 1.

The mechanisms of cytotoxicity by NK cells.

2.1.2. Genetics of KIRs

The KIR gene family includes 14 loci (KIR2DL1, KIR2DL2/3, KIR2DL4, KIR2DL5A/B, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DL1/S1, KIR3DL2, KIR3DL3 and two pseudogenes, KIR2DP1 and KIR3DP1) [31]as shown in Table 1. These loci are located on chromosome 19q13.4, which is known as the leukocyte receptor cluster (LRC); each haplotype has 9–15 KIR genes in a row [32]. Different NK cells within individuals each express a subset of the available KIR repertoire, leading to an allelic polymorphism of KIRs. Based on studies of KIR genotype variation, two major KIR haplotype groups termed the groups ‘A’ and ‘B’ are defined [33].

InhibitoryLigandsLigand missingNotes
KIR2DL1HLA‐C2 group (Cw2, C*0307,Cw4, Cw5, Cw6, C*0707, C*0709, C*1204, C*1205, Cw15, C*1602, Cw17, Cw18)HLA‐C1/C1
KIR2DL2HLA‐C1 group (Cw1, Cw3, Cw7, Cw8, Cw12, Cw13, Cw14, C*1507, C*1601/4)HLA‐C2/C2(except for C*02, *05) and HLA‐B46, B73 negative
HLA‐B46, B73
HLA‐C2(C*02, C*05)(weak interaction)
KIR2DL3HLA‐C1 group (Cw1, Cw3, Cw7, Cw8, Cw12, Cw13, Cw14, C*1507, C*1601/4)HLA‐C2/C2 and HLA‐B46, B73 negative
HLA‐B46, B73
KIR3DL1HLA‐Bw4 epitope(including HLA‐A23, A24, A32)HLA‐Bw6/Bw6 and HLA‐A23/24/32 negativeExpression level: *01502, *020 > *001, *007 >*004
KIR3DL2HLA‐A03, A11(+ EBNA peptide),HLA‐B27 dimerHLA‐A03, A11 negative, free of EBV*001 is homo‐dimer A03/A11 could not promote NK cell Licensing
KIR2DL5BUnknown
KIR2DL5TUnknown
ActivatingLigandsLigand missingNotes
KIR2DS4HLA‐A*1102,(A*1101, C*0304, C*0501)HLA‐A*1102 negative
KIR2DS1HLA‐C2HLA‐C1/C1
KIR2DS2Unknown(HLA‐C1?)
KIR3DS1Unknown(HLA‐B*2705?)
KIR2DS3/5Unknown
KIR2DL4HLA‐G
OthersLigandsLigand missingNotes
KIR2DP1Pseudogene
KIR3DP1--Pseudogene

Table 1.

Human KIRs.

Each haplotype is separated into two regions: the centromeric half (Cen) and the telomeric half (Tel). Cen and Tel motifs can be divided into Cen‐A, Cen‐B and Tel‐A, Tel‐B by the KIR genes they contain. Haplotype A is a combination of Cen‐A and Tel‐A, which consists of mainly inhibitory KIRs (KIR3DL3, 2DL3, 2DL1, 3DL1, 2DS4, 3DL2 and two pseudogenes). Other combinations are termed Haplotype B (such as Cen‐A and Tel‐B or Cen‐B and Tel‐B), composed of a large variation of genes characterized by the presence of more activating KIRs. All individuals can be categorized according to their haplotype: A/A, which is homozygous for group A haplotypes, or B/x, which contains either one (A/B) or two (B/B homozygotes) group B haplotypes[34]. As a consequence of this genetic variation, several studies report that donor‐derived NK cells can mediate the graft‐versus‐leukemia (GVL) effect or even the graft‐versus‐tumor (GVT) effect after allogeneic hematopoietic cell transplantation (HCT) [3436]. These are the results of a KIR‐ligand mismatch, the details of which are described later in this article.

2.1.3. KIRs subtypes

KIRs are type I transmembrane glycoproteins expressed on the surface of NK cells, composed of two (2D) or three (3D) extracellular Ig‐like domains and a cytoplasmic short (activating) or long (inhibitory) tail [31]. The length of the intracytoplasmic part determines the function; for example, receptors with long cytoplasmic tails with one or two ITIMs that bind to phosphatase SHP‐1, 2 allow the transduction of inhibitory signals through its dephosphorylation. In contrast, receptors with short cytoplasmic tails possess a positively charged residue (lysine) in the transmembrane domain that enables it to associate with adaptor proteins including DAP12 and process the immunoreceptor tyrosine‐based activation motifs (ITAMs) [29]. There is one exception: KIR2DL4, with a long cytoplasmic tail, binds to the activation motif of FcεRIγ and thus seems to transmit the activating signal [31].

MHC class I molecules are well‐known ligands for KIRs, but HLA‐C molecules, in particular, are the main ligands contributing greatly to NK cell activity. Polymorphisms in amino acids at positions 77 and 80 of HLA‐C show specificity for its target KIRs. Group 1 HLA‐C ligands (C1) include allele‐encoded molecules with serine and asparagine (Ser77 and Asn80), whereas group 2 ligands (C2) are characterized by allele‐encoded molecules with asparagine and lysine (Asn77 and Lys80). C1 epitopes bind specifically to KIR2DL2/3, and C2 epitopes are ligands for KIR2DL1 [37]. However, it was shown that KIR2DL2/3 might also bind to certain HLA‐C2 epitopes (C*0501, C*0202, C*0401) and some HLA‐B epitopes (HLA‐B*4601, B*7301) with very low affinity [38].

Among the inhibitory KIRs, KIR2DL1 results in a stronger inhibition compared to the KIR 2DL2/3 [38]. The third inhibitory KIR is KIR3DL1, which binds to HLA‐Bw4 epitopes and a subset of HLA‐A epitopes (A*23, A*24, and A*32). All HLA‐B have either the Bw4 or Bw6 epitope, but only the Bw4 epitope is a ligand for KIRs [39]. KIR3DL2 is a framework gene, and it recognizes HLA‐A*03 and HLA‐A*11 with a low level of inhibition [40]. Although haplotype A includes only a single activating KIR (2DL4), at least two to five activating KIR genes (2DS1, 2DS2, 2DS3, 2DS5 and 3DS1) are subject to haplotype B. However, KIR2DS1 alone is now confirmed to have matched ligand HLA‐C2 with lower avidity compared to KIR2DL1 [41].

2.2. Death Ligands

NK cells express members of the tumor necrosis factor (TNF) superfamily, the so‐called death ligands. The cognate of the receptor on target cells by these ligands on NK cells — which include TNF‐related apoptosis‐inducing ligand (TRAIL), Fas ligand (FasL) and TNF‐like weak inducer of apoptosis (TWEAK) — results in classical caspase‐dependent apoptosis [42, 43]. In the TRAIL/TRAIL receptor system, at least five receptors have been identified and two of them, TRAIL‐R1 (DR4) and TRAIL‐R2 (DR5), contain cytoplasmic death domains and are able to transduce an apoptotic signal [44]. Other TRAIL receptors — TRAIL‐R3 (DcR1) and TRAIL‐R4 (DcR2) and a soluble receptor called osteoprotegerin (OPG: TRAIL‐R5) — lack a death domain, but they are dedicated as decoy receptors to regulate TRAIL‐mediated cell death [45]. In target cell expressing TRAIL receptors, the ligation of TRAIL can lead to the activation of caspase‐8 and subsequently caspase‐3 to induce apoptosis [46, 47]. TRAIL is up‐regulated after the stimulation of interferon‐gamma (IFN‐γ) in vitro [48, 49] and also in vivo, which demonstrates that TRAIL is required for the IFN‐γ‐mediated prevention of tumors [50].

FasL is expressed on activated NK cells and cytotoxic T lymphocytes (CTLs) [42]. Although it is known that FasL is expressed on NK cells at a low level, significant amounts are stored intracellularly [51]. An activating signal of rodent NK1.1 up‐regulated the expression of FasL [52]. After FasL has bound to Fas, the Fas associated with two specific proteins, Fas‐associated death domain (FADD) and caspase‐8, to form the death‐inducing signal complex (DISC). Fas is expressed on various tissues, but the molecule is downregulated in cancers during its progression [53]. NK cells are capable of directly inducing Fas expression on tumor cells via IFN‐γ secretion, and NK cells show cytotoxicity to tumor cells expressing Fas [45].

2.3. Perforin and granzyme

Cytolytic killing using perforin and granzyme is a major mechanism in the elimination of infected cells and tumor cells by NK cells. NK cells contain cytoplasmic granules including perforin (a membrane‐disrupting protein) and granzyme (a family of serine proteases). Once NK cells recognize target cells, they form an immunological synapse, and the secretory granules fuse with the presynaptic membrane and release perforin and granzyme into the synaptic cleft. Released perforin provides transmembrane pores on the target cell and enables granzyme to diffuse into the cell. Granzyme then initiates the apoptosis of the target cells, and the NK cells detach from the dying cells and can interact with other target cells to accomplish serial killing [54]. The release of such granules stored in the NK cells is dependent on the polarization of both microtubules and actin filaments in the cytoskeleton. The increase of intracellular calcium concentration triggered by a positive balance of activating and inhibitory signals initiate the rapid move of microtubule‐organizing center (MTOC) in the cytoplasm towards the target cell, and then cytotoxic granules migrate along the MTOC [54]. Granules fuse with the presynaptic membrane, subsequently, the lytic granules can be released at the NK cell‐target cell interface[55] (Figure 2).

Figure 2.

Interaction of an NK cell with a target cell.

Advertisement

3. NK cells for clinical use

3.1. Sources of NK cells

There are several options for sources of NK cell therapy, including peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), bone marrow (BM), cell lines, human embryonic stem cells (hESCs), and induced pluripotent stem cells (iPSCs).

3.1.1. Peripheral blood mononuclear cells (PBMCs)

PBMCs are the most common source of NK cells, and PBMCs can be collected by apheresis or a specific gravity centrifugal method (e.g., Ficoll separation). However, the percentage of NK cells in PBMCs is low (5–20%), and because there is a limit on the number of cells that can be recovered from a donor by lymphocyte apheresis, it is not possible that a sufficient number of NK cells for killing the target can remain in the recipient [7]. That is, in order to stay in the recipient’s body until the target cells are killed, it is necessary to collect peripheral blood frequently, which is a heavy burden on the patient.

Numerous methods for amplification cultures of NK cells have been reported. Stem cell mobilization is a process whereby stem cells (CD34+cells) are stimulated out of the bone marrow space (e.g., the hip bones and the chest bone) into the bloodstream, and granulocyte‐colony stimulating factor (G‐CSF) is widely used as a drug for harvesting peripheral blood stem cells from patients or healthy people. Another drug, Plerixafor [56], a CXCR4 antagonist approved in 2008, has an excellent mobilization effect in combination with G‐CSF [57, 58].

An increase in a number of CD34‐positive cells harvested by mobilization reduces the number of apheresis sessions required for cell therapy, which may reduce the burden on patients. When allogeneic hematopoietic stem cells are used, GVHD in the acute phase, which is thought to be caused by T‐cell contamination, exists as a problem to be overcome. There is also a report that the induction of myeloid‐derived suppressor cells (MDSCs) by the administration of G‐CSF reduces the frequency of GVHD. It is important to note that MDSCs suppress T‐cell function through an arginine depletion by arginase‐1, the production of reactive oxygen species, and the induction of Treg cells [59, 60].

3.1.2. Bone marrow (BM)

Bone marrow aspiration removes a small amount of bone marrow fluid through a needle put into a bone under general anesthesia. Compared to apheresis, it is very rarely used as a starting material because of its high invasiveness to donors.

3.1.3. Umbilical cord blood (CB)

Hematopoietic reconstitution for Fanconi anemia treatment using cord blood was first performed in 1988 by Gluckman et al. [61]. Since then it has been widely accepted as a source of hematopoietic stem cells when autologous blood is not recommended or readily available. Cord blood (CB) can be donated to public CB banks for use by any patient worldwide for whom it is stored for potential autologous or family use. The majority of CB products used today are for hematopoietic stem cell transplantation and are accessed from public banks.

Cord blood presents no harm to the donor at the time of collection, and the frozen storage of collected samples is possible. In addition, cord blood of various blood types is classified and preserved in umbilical cord blood banks. When umbilical cord blood is used as the source of therapeutic NK cells, it is highly tissue‐compatible according to the patient’s blood type and accompanying transplantation. It is easy to select blood type‐specific umbilical cord blood, with little side effects. In addition, it is possible to minimize the risk of GVHD from the absence of T cells [6264]. As with PBMCs, a growth culture is essential because the amount of NK cells that can be obtained from a single CB sample is not an amount that can expect to provide a therapeutic effect.

3.1.4. Embryonic stem cells and induced pluripotent stem cells (ES/iPS)

Another possible source of NK cells is human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). A differentiation/expansion culture from hESCs and iPSCs to NK cells is a regimen requiring more sophisticated technology compared to the PBMCs and UCB described above. The differentiation process of human iPSCs is divided into three stages of “maintenance amplification”, “structure construction”, and “differentiation induction”. The culturing step is to generate CD34+ hematopoietic progenitor cells from hESCs and iPSCs and then differentiate these cells into NK cells.

A preclinical demonstration that NK cells can be separated from the sources of these pluripotent stem cells has been described [65, 66]. However, that demonstration enabled efficient proliferation by using mouse stromal cells as feeder cells, and the involvement of heterologous cells may limit clinical application. As with UCB, advances in the development of safe, effective and standardized clinical‐grade manufacturing protocols will provide opportunities to develop ready‐made personalization and immunogenic cell therapy.

3.1.5. Cell lines

The cell lines that have been derived from NK cells are NK‐92, NK‐YS, KHYG‐1, NKL, NKG, SNK‐6, and IMC‐1 cells [67], and several research groups are exploring the possibilities of using these cell lines for therapeutic applications. The primary advantage of an NK cell line is that it is “ready to use”, and it is possible to establish comprehensive standardization and characterization of the cell source by using the master cell bank. The cell therapy product is thus considered to be an attractive merit in manufacturing. Moreover, a more homogeneous population is obtained compared to that from peripheral blood, and its homogeneous character is another advantage when performing a genetic modification operation.

NK cell line has been applied to genetic modification technology for expressing intracellular IL‐2 for the forced expression of CD16, natural cytotoxicity receptor (NCR), chimeric antigen receptor (CAR) and NK cell activation [68, 69]. The most widely clinically used cell line is the NK‐92 cell line, which is cytotoxic to a wide range of malignant cells [70, 71]. NK‐92 cells express the receptor but hardly express KIR, NKp44 or CD16. NantKWest (Culver City, CA, USA) conducts clinical trials using NK‐92 cells (Neukoplast™) and has completed a Phase 1 study (U. S. National Clinical Trial [NCT] #00900809 and NCT #00990717). Moreover, the company has begun phase 1 and phase 2 trials of haNK (high‐affinity NK cells) [72] engineered to express CD16 (NCT #02465957 and NCT #03027128). In addition, as another attempt, the development of CAR‐TNK expressing CD7 or CD33 has been advanced (NCT #02944162 and NCT #02742727).

3.2. Manufacturing method

The number of NK cells contained in a collectable amount of UCB or peripheral blood is not enough to achieve a clinical therapeutic effect. A long‐term culture method is necessary to overcome this problem. For starting material, T cells and/or B cells are removed with magnetic beads to increase the purity of NK cells. This is also to prevent the proliferation of T cells caused by IL‐2 during the culture period of NK cells and to avoid lower purity of the final product [8, 73].

In addition to the important cytokine IL‐2, there are IL‐15, which is necessary for both the maturation and survival of NK cells [21]. IL‐2 and IL‐15 share the same receptor component IL‐2/15Rβ and a common γ chain, and they are used in a culture method without the use of feeder cells [74, 75]. IL‐21 [76], a member of the IL‐2 cytokine family, is a potent immunostimulatory cytokine that shows diverse regulatory effects on NK cells, T cells and B cells [77, 78] and also has the effect of enhancing rituximab‐mediated antibody‐dependent cell mediated cytotoxicity (ADCC) of mantle cell lymphoma [79]. In addition, there is a culture method using feeder cells to efficiently expand NK cells ex vivo. As feeder cells, monocytes, irradiated PBMCs, the K562 cell line and a genetically modified cell line are used.

For example, there are systems using a co‐culture with NK cells and monocytes [80], with CB CD34+ cells and bone marrow stromal cells [81], or K562 cells transfected with IL‐21 [82]. There is also a completely closed culture using Epstein‐Barr virus‐transformed lymphoblastoid cell lines [83]. Thus, the use of feeder cells is an important method for securing a number of cells that can be expected to have a therapeutic effect. However, because of the problem of infectious disease risk presented by the use of an allogeneic feeder, the regulatory hurdles in the manufacture of pharmaceutical products are high [84].

To overcome this problem, Yonemitsu et al. reported a method of culturing highly activated NK cells with ≥90% purity from PBMCs in a completely closed and feeder‐free system under the good manufacturing practices (GMP) [13]. As another feeder‐free culture method, Spanholtz and colleagues reported a culture method that achieved amplification efficiency and high purity of 10,000 times or more in 6 weeks from UCB CD34+ hematopoietic stem cell (HSCs), using a closed system process based on GMP [85]. Knorr and colleagues reported differentiation induction from CD34+ hematopoietic progenitor cells produced under feeder‐free conditions to cytotoxic NK cells [86].

3.2.1. NK cell‐based immunotherapy: autologous cells

Clinical trials using autologous NK cells have been performed targeting solid tumors such as colorectal cancer, non‐small cell lung cancer, melanoma, kidney cancer and esophageal cancer [8789]. In general, autologous NK cell therapy is safe without side effects such as GvHD [87], but its therapeutic effect is limited to some cancer types [74]. An activation culture with IL‐2 and OKT3 or Hsp70 has been reported to be able to efficiently induce the proliferation of NK cells [74], in particular, the retronectin culture method of Sakamoto et al. showed a high amplification efficiency (about 4720‐fold) [89].

3.2.2. NK cell‐based immunotherapy: allogeneic cells

Allogeneic NK cell products are used for the treatment of malignant tumors such as leukemia, renal cell carcinoma, colorectal cancer, and lymphoma. The major risk of allogeneic NK cell transplantation is the onset of GvHD. Measures against GvHD include the use of immunosuppressive agents, injection of high‐purity NK cells by CD3 depletion, and the selection of donors consistent with the host HLA [74, 75]. In the case of haploidentical donors and recipients, to avoid GvHD, it is necessary to strictly perform T‐cell depletion. In many studies, CD56+ is enriched after the removal of CD3+ T cells [17, 90, 91]. In cases of an allogeneic type, HLA typing and confirmation of KIR by flow cytometry are carried out, particularly in order to select the optimum donor. For details on the selection criteria, please refer to a later section of this article.

3.2.3. Synergistic effect: antibody drugs

An antibody drug is, in short, a medicine that functions based on the specificity with which an antibody recognizes an antigen. The characteristics of antibody drugs are high specificity (low toxicity) and high stability in vivo. The antibody binds only to the target antigen, and not to any other, which leads to the intended medicinal effect with only rare unexpected side effects. Antibodies are present at a stable level in the blood, and antibody medicines can also be detected at a stable level in the blood for a long period after administration, and they can exert their medicinal effects over a long term. More than 50 antibody drugs have been approved in Japan, the U.S. and Europe. Target diseases include cancer, rheumatoid arthritis and psoriasis, but most of the targets are cancers. One of the action mechanisms of antibody drugs used to treat cancer is ADCC, in which NK cell plays a central role. Table 2 shows FDA‐approved antibodies for ADCC to treat cancer.

Drug nameTrade nameTypeTargetCancersApproved year
RituximabRituxanChimeric IgG1κCD20Non‐Hodgkin)s B‐cell lymphomas Chronic lymphocytic leukemia1997
MabThera
TrastuzumabHerceptinHumanized IgG1κHER2Adenocarcinoma of the stomach or gastroesophageal junction1998
Breast cancer
AlemtuzumabCampathHumanized IgG1κCD52B‐cell chronic lymphocytic leukemia2001
CetuximabErbituxChimeric IgG1κEGFRColorectal cancer2004
Head and neck cancer
OfatumumabArzerraHuman IgG1κCD20Chronic lymphocytic leukemia2009
PertuzumabPerjetaHumanized IgG1κHER2Breast cancer2012
ObinutuzumabGazyvaHumanized IgG1κCD20Chronic lymphocytic leukemia2013
Follicular lymphoma
DinutuximabUnituxinChimeric IgG1κGD2Neuroblastoma2015
DaratumumabDarzalexHumanized IgG1κCD38Multiple myeloma2015
ElotuzumabEmplicitiHumanized IgG1κSLAMF7Multiple myeloma2015

Table 2.

FDA‐approved antibodies for ADCC to treat cancer.

ADCC mediated by NK cells begins with the recognition of antibodies bound to target cells. NK cells express two Fc receptors, CD16a (FcγRIIIa) [92] and CD32c (FcγRIIc) [93]. These Fc receptors recognize and bind to IgG1 and IgG3 and have a high affinity for IgG3 [94]. NK cells that recognize the antibody on the target cells transmit their signals intracellularly and kill the cells.

The signaling of human CD16 is mediated via FcεRIγ, CD3ζ, or FcεRIγ‐CD3ζ heterodimer. These molecules contain an ITAM and are phosphorylated when the antibody binds to CD16 [42]. CD32c (FcγRIIc) contains an ITAM‐like sequence in the cytoplasmic domain (which is similar to that of FcγRIIa), suggesting that it transmits a signal via SRC‐SYK (the SRC family of kinases and spleen tyrosine kinase [SYK]) signaling pathways [95]. However, the expression of CD32c was less than half of that of NK cells [96, 97]. Many studies focusing on CD16 have thus been conducted.

Several genetic polymorphisms of CD16 exist. Among them, the amino acid at position 158 has been shown to be important for the strength of the affinity for antibodies. The affinity depends on whether the amino acid at position 158 is phenylalanine or valine, and the valine type (158 V) has a higher affinity for the Fc of IgG. A number of reports have indicated that differences in the affinity for antibodies are correlated with therapeutic effects, and many studies have analyzed the clinical responsiveness of this gene polymorphism and antibody therapy. Cartron et al. examined the effects of rituximab treatment for non‐Hodgkin’s lymphoma, and they reported a higher objective response rate in CD16 (158 V) homozygous patients compared to CD16 (158F) carrier patients [98]. Wang et al. analyzed the outcomes of rituximab treatment for Follicular Lymphoma and reported significantly more likely progression‐free survival at 2 years in CD16 (158 V) homozygous patients compared to CD16 (158F) carrier patients, at 45 and 14%, respectively [99].

These results suggest that the affinity of CD16 for antibodies correlates with the therapeutic effect. Thus, focusing on CD16, the modification of NK cells has been attempted. Binyamin et al. reported that introducing CD16 (158 V) into NK‐92 cells not expressing CD16 improved the cytotoxicity against B‐cell lymphoma with rituximab [100]. Carlsten et al. reported high ADCC activity of cultured NK cells from healthy donors with CD16 (158F/F) and transduced CD16 (158 V) mRNA by electroporation against rituximab‐coated CD20+ B‐cell lymphoma cells [101].

As described above, since NK cells mediate antibody‐dependent cytotoxic activity via Fc receptors, compatibility with antibody drugs targeting ADCC is desirable. As a strategy to further augment the antitumor effect, a plausible strategy is to enhance the affinity between the Fc receptor and the antibody. Low‐molecular‐weight compounds that are able to inhibit the shedding of CD16 have been reported. It is known that CD16 is cleaved by a protease such as a disintegrin and a metalloprotease 17 (ADAM17) when cells are activated [102], and thus in order to exert more sustained and enhanced ADCC activity, a method of inhibiting the cleavage of CD16 on NK cells may be important. In fact, inhibitors of ADAM17 enhanced the activity of NK cells [102, 103]. Another method to inhibit the cleavage of CD16 is a genetic modification. The substitution of the serine residue at position 197 in CD16 by a proline prevents the cleavage of CD16 on NK cells [104]. It may be possible to promote the antitumor effect by using a CD16 mutant.

Advertisement

4. Optimized selection of patients/donors

4.1. Clinical outcomes: on the KIR ligand mismatch model

Research reports focusing on the KIR of NK cells and the HLA of tumor cells for the purpose of treating leukemia have been drawing attention since the 2000′s. In the report by Ruggeri et al. published in Science in 2002, the cases in which HLA‐ABC recognizable by NK cells is present in a donor but not in a recipient were defined as “KIR mismatch”. It was speculated that in such cases, NK cells not receiving suppression signals from the donor HLA attack the cells of the recipient. When Ruggeri et al. started this research, it was not known that NK cells are a heterogeneous population expressing multiple inhibitory receptors, or that the specific antibodies that can be used for analysis are inadequate. Ruggeri and colleagues, therefore, examined only the HLA of donors and recipients without examining the KIR, and they analyzed patients divided into the two groups of HLA matched and mismatched transplantations. Although it may not have been an appropriate observation based on the current knowledge, at that time, the analysis was based on the following basic research data.

Ruggeri et al. examined whether CD56‐positive NK cell clones collected from donors could attack leukemia cells of the recipient. They found that NK cell clones that attack leukemia cells exist, and their high frequency (≥2%) correlates well with the cases in which the donor’s and recipient’s KIR ligand (HLA) do not match in the GVH direction. Ruggeri et al. analyzed 35 acute lymphoblastic leukemia (ALL) and 57 acute myeloid leukemia (AML) patients who had allogeneic hematopoietic stem cell (HSC) transplantations at their institution. The result was a breakthrough result in 20 AML patients who were not KIR ligand mismatches in the GVH direction, with 0% relapse after transplantation [105]. This report was the subject of much attention, and then it was decided to conduct data analyses in medical institutions around the world seeking reproducibility of the results.

The analysis by Ruggeri et al. was by the method known as the “KIR ligand mismatch model”, which examines only the donor and recipient HLA‐ABC without checking the KIR. This method determines whether a KIR match or mismatch is determined. Researchers all over the world could thus easily use this method. However, most of the analysis results obtained in this way conflicted with the report by Ruggeri et al.

As an example, we will describe the analysis results obtained from the study of the Japan Marrow Donor Program (JMDP). In 2007, Morishima et al. reported the results of their analysis of 1790 patients who underwent an allogeneic bone marrow transplantation in accord with the KIR ligand mismatch model [106]. In conventional domestic allogeneic HSC transplantation, the criterion for donor selection is that the HLA‐AB and ‐DR matched, and by this criterion 534 of the 1790 cases were HLA‐C mismatched. As a result, the overall survival rate, the recurrence rate, and the incidence of GvHD were poor in the group in which the KIR ligand HLA‐C was mismatched in the GVH direction.

The finding that allogeneic HSC transplantation has no merit in KIR mismatches is common in the report about cases from the U.S. National Marrow Donor Program (NMDP) and the European group for blood and marrow transplantation (EBMT) [107]. Comparing Ruggeri’s 2002 report with Morishima’s 2007 report, the biggest difference was the cell source for transplantations, the former being CD34+ cell transplantation from haploidentical donors and the latter being conventional bone marrow transplantation. In addition, Ruggeri et al. used anti‐thymocyte globulin (ATG) in all cases. A re‐analysis of the registered JMDP cases reconfirmed that the transplantation performance of the HLA‐C matched group was also good and that of the group with the HLA‐C mismatch was poor [108].

Interestingly, the disadvantage of this HLA‐C mismatch was not observed in the ATG administration group. In other words, it is suggested that not NK cell dysfunction but T cells induced by HLA‐C mismatch had exacerbated the transplantation results. T cell‐depleted grafting and ATG might avoid the T‐cell response. There are certain reports that HLA‐C mismatches are recognized by cytotoxic T lymphocytes (CTLs). There are two papers that reported a total of nine CTL clones from two patients who developed GvHD [109, 110]. Interestingly, all of the targets of the nine CTL clones were HLA‐C, which was different from the HLA‐C of the recipient. There is no doubt that the difference in HLA‐C could be a target for CTL.

4.2. Receptor ligand model or missing ligand model

Although the KIR ligand mismatch model investigated only ligand (i.e., HLA) differences, eventually some researchers noted that donors’ KIR should also be examined and analysed by different approaches were developed. In 2004, Leung et al. analyzed 36 children who received selective CD34+ cell transplantation from haploidentical donors [111]. They first examined the presence or absence of inhibitory KIR in donor cells by flow cytometry (i.e., a phenotype assay). It was speculated that the recipient would have a KIR mismatch if the recipient did not have a ligand for KIR (suppressing type) of the donor. This is the “receptor‐ligand model”.

For example, when the donor has KIR2DL1 with HLA‐C1/C1, when the recipient is HLA‐C1/C1, it is considered a KIR match in the KIR ligand mismatch model, but it is a ligand mismatch of KIR2DL1 in the receptor‐ligand model. This is a very frequent combination for Japanese (HLA‐C1/C1 = 85%, KIR2DL1+ = 99%). Conversely, if the donor does not have KIR2DL1 with HLA‐C1/C2, if the recipient is HLA‐C1/C1, it is considered a KIR mismatch in the KIR ligand mismatch model, but it is considered a KIR match in the receptor‐ligand model. However, since there are few KIR2DL1 and HLA‐C1/C2 among Japanese, this combination is extremely rare.

An analysis conducted to determine which model can predict recurrence more accurately revealed that the receptor‐ligand model is superior [111]. Hsu et al. also reported their analysis based on the receptor‐ligand model [112]. They investigated 178 cases of T cell‐depleted transplantation and found that the positive rates of KIR2DL1, KIR2DL2/3, and KIR3DL1 were 93, 99, and 92%, respectively, in the donor gene, and that although there were 112 cases (63%) of the 178 patients who were expected to exhibit the GVL effect, there was a significant difference in the recurrence rate, and the disease‐free survival rate/overall survival rate was also good in the AML and myelodysplastic syndrome (MDS) patients.

Hsu et al. presented a new idea in 2006 [113]. It is hard to examine the donor’s KIR by genetic testing, but donors usually have genes of KIR2DL1, KIR2DL2/3, and KIR3DL1. Therefore, HLA‐C1/C1 homozygous, HLA‐C2/C2 homo and HLA‐Bw6/Bw6 homo patients identified by examining only the recipient’s HLA may experience the GVL effect from donor NK cells. This method is called the “missing ligand model”. It was designed for transplantation performance analyses, not for donor selection. Hsu et al. analyzed 1770 patients who had allogeneic (unrelated) T‐cell‐depleted transplantation. A University of Minnesota study examined 2062 cases from the U.S. NMDP [114], and based on the idea that NK cells have a GVL effect, they excluded 568 Japanese subjects or analyzed only partial diseases with good prognosis. Nonetheless, that paper is very interesting as it shows the difference in the distribution of KIR ligand between Japanese and Westerners and the difference in the recurrence rate of disease [113]. We can see how Japanese are ‘biased’ toward HLA‐C1.

Although few cases of GvHD in Japanese have been reported, the recurrence rate of HLA‐C1/C1 patients is not much different from that of Westerners. In addition, among the Japanese, the recurrence rate is extremely small when the recipients are HLA‐C2/C2 homozygous, which is a minority. However, caution is required in the interpretation of the results, as there are only three cases in which the recipient had HLA‐C2/C2. Further detailed analysis is expected in the future.

4.3. Therapeutic effects of allogeneic transplantation of NK cells, and their limitations

In 2007, Ruggeri et al. reported the results of 122 cases of AML [115], which included 57 cases [105] and the other 55 cases. In the KIR ligand mismatch model, there were 51 cases of KIR mismatch in the GVH direction and 61 cases of matches, and the number of remission cases at the time of transplantation and the cases which were refractory to treatment were approximately 50% of the cases. The results showed that although the relapse rate was significantly lower in the cases of remission at the time of transplantation in the KIR mismatch in the GVH direction (3 vs. 47%, p = 0.003), in the cases in which the treatment was refractory, no treatment effect was observed (32 vs. 37%, p = NS).

Is it true that NK cells exert a GVL effect? A direct answer to that question has been reported as a KIR‐mismatch NK cell transplantation in recent years. Ten children with AML [7] and 13 adults with AML [116] underwent the transplantation of CD56‐positive NK cells harvested from haploidentical donors. The collected NK cells were considered KIR mismatches, and in both studies, only NK cells were transplanted after pretreatment (fludarabine and cyclophosphamide). The number of transplanted NK cells was 29 × 106/kg for the children and 2.7 × 106/kg for the adults; the numbers of transplanted T cells were <1 × 103/kg and < 1 × 105/kg, respectively. After transplantation, 1–10 million units of IL‐2 were administered every other day, 6 times.

As a result, transient engraftment of donor NK cells was observed in all cases. Donor‐derived NK cells occupied 7% (1–30%) of the peripheral blood lymphocytes at the peak of day 14 in the children’s report, and at day 28, donor‐derived NK cells have been detected from three of 10 patients [7]. All of the child patients had AML in complete remission, and all cases did not recur. Among the adult patients, whose leukemia was worse, one of five patients with clear recurrence showed transient remission, and two patients with genetic recurrence AML were remitted [116]. From the above results, it was demonstrated that NK cells exert a sufficient antitumor effect if the residual tumor is relatively small. However, it is not yet clear whether the therapeutic effect is proportional to the number of NK cells administered. It should be noted that even though transplanted CD3‐positive cells are limited to ≤1 × 103/kg, there was no case of GvHD onset in either group.

4.4. Selection of KIR in allogeneic hematopoietic stem cell transplant donors

The target diseases in which the therapeutic effect by NK cells is confirmed in analyses of various clinical tests are mostly limited to AML. Clinical trials using conventional NK cells often do not even target other diseases. There are reports that NK cells do not show a therapeutic effect against ALL because of lymphoid cells highly MHC class I, and therefore, the inhibitory signal is strong [117]. In addition, KIR ligand mismatch can lead to GvHD by CTLs as described above. In order to avoid this, ingenuity such as umbilical cord blood transplantation or the use of ATG may be necessary.

4.4.1. Donor selection based on inhibitory receptors

There is a reason to expect the GVL effect in donor NK cells not receiving a KIR signal from the recipient HLA. The main inhibitory receptors are KIR2DL1, KIR2DL2/3, KIR3DL1, and KIR3DL2. There is no evidence that the effects of these four inhibitory receptors are identical. Particularly with regard to KIR3DL2, there are reports that the KIR‐positive NK cells are not even licensed, even if the recipient has HLA‐A3 or A11 (KIR3DL2 ligands) [118]. Most NK cell transplantations have been expected to provide a therapeutic effect due to missing self of KIR2DL1 or KIR2DL2/3, and the number of transplantations in which a therapeutic effect by missing self of KIR3DL1 have been expected is very low. There is no transplantation in which the therapeutic effect by missing self of KIR3DL2 alone is expected.

For example, if the recipient is HLA‐C1/C1 if the KIR ligand mismatch model is used, an HLA‐C1/C2 or HLA‐C2/C2 donor would be selected. Even if transplantation is assessed using the receptor‐ligand model, the donor’s KIR is often unknown. However, in Japanese, most (approx. 99%) donors KIR2DL1 can be considered positive. If so, in the receptor‐ligand model, it seems that any donor could be chosen, but NK cells derived from an HLA‐C1/C1 donor may show a lower GVL effect. When the recipient is HLA‐Bw6/Bw6, it must first be confirmed that all of the HLA‐A23, ‐24 and ‐32 are negative, and then an HLA‐Bw4‐positive donor should be selected. However, since 7% of Japanese are negative for KIR3DL1 gene, it must be confirmed that KIR3DL1 is positive for the donor by flow cytometry, or that the genotype is either KIR3DL1*01502 or KIR3DL1*020.

4.4.2. Donor selection based on haplotypes

It was reported that donors should be chosen for haplotype B when considering the activating receptors [34]. In that study, Cooley et al. analyzed 448 AML cases of NMDP, and they observed that the 3‐year overall survival rate when the donor was haplotype BX was 31%, significantly higher than the rate of 20% when it was haplotype AA (p < 0.01). In addition, the recurrence decreases the most in cases that the donor had Cen‐B/Cen‐B, and it is reported that Tel‐B also leads to a reduction of recurrence and improvement of prognosis [119].

Based on the data from three groups in the United States (Memphis, Sloan Kettering, Minnesota), Leung et al. advocated a donor selection algorithm for NK cells [120]; in the transplantation of T cells containing bone marrow or peripheral blood stem cells, donors should be HLA matches, and donors with KIR ligand mismatch should be avoided. Conversely, in T cell‐depleted transplantation or umbilical cord blood transplantation, a donor for a KIR ligand mismatch should be chosen. It is also recommended that a donor with KIR that can attack the recipient’s HLA (possibly KIR haplotype B) be selected. In Japan as well, if KIR haplotype testing of the donor banks becomes possible in the future, or if the NK cell preparation is put to clinical use, it will be possible to test this algorithm described by Leung et al.

4.4.3. Donor selection based on activating receptor

KIR2DS1 is activated by HLA‐C2. This means that if the recipient is positive for HLA‐C2 and the donor is KIR2DS1‐positive (Tel‐B = haplotype BX), KIR2DS1‐positive NK cells will be activated and will kill the recipient’s tumor cells. In actual transplantation, when NK cells were cultured in vitro to examine the antitumor effect on leukemic cells of patients, KIR2DS1‐positive NK cells killed tumor cells of recipients with HLA‐C2 [121]. An analysis of the KIR gene of AML donors and recipients (1277 cases) from the U. S. NMDP and the Center for International blood and marrow transplant research (CIBMTR) provided large‐scale proof using actual transplantations, and the report was published in The New England Journal of Medicine [122]. The analysis revealed that recurrence was significantly reduced when the donor had KIR2DS1 (26.5 vs. 32.5% (KIR2DS1‐negative), p = 0.02). However, this effect was canceled when the donor was HLA‐C2/C2. This is thought to be a disarming phenomenon, and it can be explained as follows: the HLA‐C2/C2‐derived KIR2DS1‐positive clone is inactivated.

In that report [122], the proportion of donors with the KIR2DS1 gene was 33%. In the report by Yabe et al. [108], the KIR2DS1 gene‐positive rate was 38%. Since Yabe et al. focused only on recipients of HLA‐C2/C2, they did not analyze KIR2DS1‐positive donors because the number of cases was too small. In addition, it was reported that when the donors had KIR3DS1, although a decrease in the recurrence rate was not observed, the mortality rate decreased slightly [122]. Yabe et al. also analyzed leukemia patients and reported that transplantation from KIR3DS1 donors reduced the rate of acute GvHD [123]. It was also reported that the likelihood of acute GvHD increases when the donor is haplotype AA [124, 125]. The mechanisms underlying KIR/HLA interactions remain unclear, but these reports may be a reference for donor selection.

4.5. Immune checkpoint inhibitors

4.5.1. Checkpoint of NK cells

Checkpoint inhibitors are an extremely promising approach among immunotherapies. Treatment with anti‐CTLA4 or anti‐PD‐1 antibody restored the T‐cell activity in cancer patients and resulted in tumor regression in several patients. A combination of both of the checkpoint inhibitors anti‐CTLA4 and anti‐PD‐1 could further enhance therapeutic benefits [126, 127]. It has been shown that NK cells from patients with multiple myeloma and renal cancer express PD‐1, the signal of which reduce the cytolytic activities of NK cells [128, 129]. Treatment using patient‐derived PD‐1+ NK cells with the anti‐PD‐1 antibody (pidilizumab, CT‐011) increased the NK cell‐mediated killing of autologous cancer cells in vitro [128]. The therapeutic benefit of activating PD‐1+ NK cells in cancer patients is currently not well understood, and the major therapeutic effect is certainly due to the re‐activation of exhausted T cells.

4.5.2. Combination with a checkpoint inhibitor: expansion of the therapeutic spectrum

A loss or down‐regulation of HLA class I antigens in tumor cells has been frequently observed in a variety of human malignancies, and this represents an important cancer‐immune escape mechanism [130134]. Using a panel of monoclonal antibodies on tumor tissue sections, these loss or down‐regulation has been found in 60–90% of tumors [135140]. Early studies using immunohistological analyses of different tumors showed a very low frequency of allelic loss. However, with the arrival of other techniques, such as studies of microsatellites to detect the loss of heterozygosity (LOH) on chromosome 6, it has been shown that LOH (haplotype loss) is the most frequent alteration of HLA class I expression [139, 141144]. This alteration is caused by various defects in the HLA genomic region (i.e., the short arm of chromosome 6, 6p21), including chromosomal dysfunction, mitotic recombination, and genetic conversion.

The nature of the antigens that allow the immune system to distinguish cancer cells from noncancer cells has long remained obscure. Recent technological innovations have made it possible to dissect the immune response to patient‐specific neoantigens that arise as a consequence of tumor‐specific mutations, and emerging data suggest that the recognition of such neoantigens is a major factor in the activity of clinical immunotherapies. These observations indicate that the neoantigen load may form a biomarker in cancer immunotherapy and provide an incentive for the development of novel therapeutic approaches that selectively enhance T‐cell reactivity against this class of antigens.

If there is a neoantigen that can be a target of CTL and the patient has MHC on which the antigen is presented, and if the MHC is not lost from the tumor cells, treatments using CTL as an effector (e.g., checkpoint inhibitors) may be effective. NK cells that preferentially kill tumor cells whose expression of MHC has decreased by MHC non‐restriction are expected to have a synergistic effect with a checkpoint inhibitor.

Advertisement

5. Future directions

5.1. Genetic modification–Gene transfer to NK cells

In order to genetically modify NK cells, efficient methods for gene transduction into NK cells are necessary. To date, such methods include viral transduction, electroporation, and nucleofection [145]. Gene transfer to not only NK cell lines such as NK‐92, but also primary NK cells has been conducted. Although various gene transfer efficiencies have been described, their transduction efficiency into NK cells is generally not high. Mainly retroviral vectors and lentiviral vectors are used for gene transfer. Since a retrovirus vector cannot transduce genes into non‐dividing cells, such a vector is suitable for use with NK cell lines such as NK‐92 cells. When a retrovirus vector is used for primary NK cells, it is necessary to amplify the NK cells, and the transduction timing is important.

In contrast, lentiviral vectors are capable of gene transfer into both dividing and non‐dividing cells. The lentiviral vectors RD114, 10A1, GALV, and VSV‐G are used for the envelope of the viral vector, and RD114 and VSV‐G seem to be suitable. However, it has been reported that viral vectors are recognized by antiviral mechanisms such as intracellular pattern recognition receptors, and apoptosis is induced [146]. The introduction efficiency was therefore not high.

Efforts are underway to improve the transduction efficiency. For example, by using a cytokine combination (e.g., IL‐2 + IL‐15 or IL‐2 + IL‐21), the transduction efficiency into NK cells by VSV‐G pseudotyped lentiviral vector was improved by approx. fivefold compared to single cytokine‐stimulated NK cells [146]. In addition, inhibitors of intracellular antiviral responses were evaluated, and the results indicated that BX795 (an inhibitor of the TBK1/IKKe complex) improved the transduction efficiency by approx. 10‐fold [146]. Guven et al. reported transduction into 75.4% of NK cells after 21 days of culture by a two‐round transduction with the GALV‐pseudotyped retroviral vector [147]. Further improvement of the transduction efficiency into NK cells by using viral vectors is desired.

In non‐viral gene transfer, mainly electroporation has been studied. Electroporation is a method of physically pulling a minute hole in a cell membrane by applying an electric pulse to a cell suspension and sending the nucleic acids into the cells. The transfer of genes into NK cells using this method has been performed. High transfection efficiency and a high survival rate have been reported in both NK cell lines and primary NK cells. Much higher transfection efficiency was achieved using mRNA compared to using DNA [148152]. Introduction by nucleofection has also been tried, but the efficiency was not high [153, 154].

In light of these reports, the transfection of mRNA by electroporation is considered to be an efficient method from the viewpoint of gene transfer into NK cells. Shimasaki and colleagues reported that NK cells transfected with mRNA encoding CD19‐specific CAR (anti‐CD19‐4‐1BB‐CD3ζ) by electroporation showed enhanced cytotoxicity for tumors in animal models [154]. On the other hand, the transfection of mRNA can obtain transient gene expression. Further research is required to determine whether a treatment effect can be expected. When persistent gene expression is the goal, gene transfer by retroviral or lentiviral vectors is suitable.

It is necessary to select a suitable gene transfer method for the purpose of treatment. In any case, it is desirable to develop more efficient, simple, highly reproducible and clinically applicable gene transfer methods for NK cells.

5.2. Improved persistence of NK cells in vivo

As a strategy for the genetic modification of NK cells to improve their survival in vivo, methods for transducing cytokines such as IL‐2 and IL‐15 into NK cells have been reported, since it was demonstrated that a local administration of IL‐2 resulted in enhanced functioning of NK cells [155, 156]. IL‐15 has already been used in patients with metastatic melanoma and metastatic renal cell carcinoma, and it has been reported to induce the proliferation and clinical response of NK cells. Following these findings, attempts to introduce IL‐2 and IL‐15 genes into NK cells were reported. Nagashima et al. showed that the NK‐92 cell line transduced with IL‐2 genes with a retroviral vector propagated for >5 months irrespective of IL‐2 and showed higher antitumor activity than the parental cell line in vitro and in vivo [157]. Imamura et al. showed that the transfection of membrane‐bound IL‐15 into human PBMC‐derived NK cells using a retroviral vector resulted in the cells’ survival for 2 months without the addition of cytokines in vitro, and that the cytotoxicity of the transduced NK cells was enhanced; moreover, the antitumor activity was observed in a mouse model [158].

Sahm et al. transduced IL‐15 gene into NK‐92 cells and observed cell proliferation in the absence of cytokines. They also showed that the co‐transduction of an EpCAM‐specific chimeric antigen receptor and IL‐15 into NK‐92 cells enhanced the specific and efficient cytotoxic activity [159]. Jiang et al. reported high cytotoxic activity of a human NK cell line (NKL) transduced with IL‐15 in vitro against human hepatocellular carcinoma; the transduced NKL suppressed tumor growth and prolonged survival in human hepatocellular carcinoma‐transplanted model mice [160]. These reports suggested that NK cells transduced IL‐2 or IL‐15 can proliferate sustainably in vitro and in vivo, which resulted in improved antitumor activity.

5.3. Improvement of homing

It was reported that NK cells expressed different chemokine receptors depending on the activating state [161, 162]. Proper homing to the tumor tissue is an important factor in eliciting the antitumor activity of NK cells. Carlsten et al. showed that ex vivo expanded NK cells derived from PBMCs transfected with CCR7 mRNA using electroporation migrated significantly to CCL19, a ligand of CCR7 [163]. Sonamshi et al. reported that NK cells transferred CCR7 protein from feeder cells using trogocytosis, but not genetic manipulation, promoted the migration to CCL19 and CCL21 in vitro, and that the NK cells were transferred to lymph nodes in a mouse model [101]. There are few reports of genetic modification targeting homing receptors, but further progress in this field is expected.

5.4. Improvement of tumor‐specific cytotoxic activity

As an approach to enhance specificity to tumor cells, a technique using a chimeric antigen receptor (CAR) should be mentioned. CAR is a chimeric protein composed of a single‐chain antibody (the antigen‐specific binding site) in which a light chain and a heavy chain derived from a monoclonal antibody recognize a tumor cell surface antigen, a transmembrane domain, and an intracellular signal domain.

In the first‐generation CARs, the intracytoplasmic signal domain is composed of only the CD3ζchain. In addition to CD3ζ, the second‐generation CARs have another T cell costimulatory signal domain (CD28, 4–1BB, OX40, etc.), and the activation signal is efficiently transmitted. In the third‐generation CARs, two or more costimulatory signal domains are inserted. CAR‐introduced T cells have already been used clinically, and excellent results have been obtained in some cases [164167]. One of the problems with CAR‐T cell therapy is a serious side effect. CAR‐T cells attack not only target cells but also non‐target normal cells. For example, CD‐19 CAR‐T cells kill not only tumor cells but also normal B cells, which cause B‐cell deficiency [168]. In HER2‐specific CAR therapy for colorectal cancer with lung metastasis, death due to a pulmonary complication accompanied by high cytokinemia has been reported [169].

NK cells are an attractive alternative to T cells, as NK cells have the following advantages. HLA matching is not necessary, and NK cells are used as allogenic cells. Their lifespan is limited, and they can be expected to be excluded from the body before severe side effects occur, after they kill cancer cells. The reported CAR‐NK cells are summarized in Table 3. NK cells transduced with CD19‐CAR or CD20‐CAR have been used when targeting B‐cell malignancies. Bossel et al. showed that NK‐92 cells transfected with anti‐CD19‐CAR by using electroporation had high cytotoxic activity against a CD19+ cell line and primary chronic lymphocytic leukemia (CLL) [150]. They also showed that tumor cells can be eliminated in xenograft mouse models by using NK‐92 cells transduced with anti‐CD19 CAR with lentiviral vectors [170]. Imai et al. also showed that primary NK cells transduced with anti‐CD19–CD3ζ with a retroviral vector killed CD19+ cells and that the cytotoxicity was improved by adding a signaling domain of 4–1BB to anti‐CD19–CD3ζ [171].

NK cell typeTarget antigenCancerCo‐stimulatory domainGene transfer methodRef
NK‐92 cell lineErbB2 (HER‐2)Breast, ovarian carcinomaCD3ζRetrovirus[175]
NK‐92 cell lineCD20B‐ALL, CLLCD3ζRetrovirus[170]
NK‐92 cell lineCD19B‐ALL, CLLCD3ζElectroporation (mRNA)[151]
NK‐92 cell lineEpCAMBreast carcinomaCD28/CD3ζLentivirus[160]
NK‐92 cell lineHLA‐A2 EBNA3CEBV+ T‐cell LymphomaCD28/CD3ζRetrovirus[178]
NK‐92 cell lineGD2NeuroblastomaCD3ζRetrovirus[179]
NK‐92 cell lineCD19/CD20B‐ALL, CLLCD3ζElectroporation (plasmid DNA)[180]
Lentivirus
NK‐92 cell lineCD19/CD20B‐ALL, CLLCD3ζLentivirus[68]
NK‐92 cell lineCS1Multiple myeloma.CD28/CD3ζLentivirus[181]
NK‐92 cell lineCD138Multiple myelomaCD3ζLentivirus[182]
NK‐92 cell lineErbB2 (HER‐2)Breast carcinomaCD28/CD3Electroporation (plasmid DNA)[176]
NK‐92 cell lineErbB2 (HER‐2)Breast, ovarian and renal cell carcinomaCD3ζLentivirus[183]
CD28/CD3ζ
CD137/CD3ζ
YTS cell linePSCAProstate cancerCD3ζLentivirus[184]
DAP12 TM and signaling
YT cell lineCD33AMLCD3ζElectroporation (plasmid DNA)[150]
YT cell lineCEAColon carcinomaCD3ζElectroporation (plasmid DNA)[148]
PBMC‐NKCD19B‐ALL, CLLCD3ζRetrovirus[171]
DAP10
CD137/CD3ζ
PBMC‐NKErbB2 (HER‐2)Breast, ovarian and renal cell carcinomaCD28/CD3ζRetrovirus[174]
PBMC‐NKCD19/GD2CD3ζ or 2B4 aloneRetrovirus[185]
2B4/CD3ζ
CD137/CD3ζ
PBMC‐NKCD19B‐ALL, CLLCD137/CD3ζElectroporation(mRNA)[152]
PBMC‐NKCD20B‐ALL, CLLCD137/CD3ζElectroporation(mRNA)[186]
PBMC‐NKNKG2D ligandswide rangeCD3ζ (with DAP10)Retrovirus[177]
Electroporation(mRNA)

Table 3.

CAR‐NK cells.

The gene human epidermal growth factor receptor 2 (HER2) is overexpressed in many breast cancers and is correlated with disease progression [172, 173]. It is therefore considered one of the suitable targets of CAR. Kruschinski et al. transduced anti‐HER2–CD28‐CD3ζ into human primary NK cells using a retroviral vector, and the results demonstrated cytotoxicity to an HER2+ cell line. This cytotoxicity was correlated with the HER2 expression level on target cells [174]. Uherek et al. reported that anti‐HER2–CD3ζ‐CAR retrovirally transduced NK‐92 cells efficiently killed cell lines derived from ErbB2‐positive breast carcinoma, ovarian carcinoma, and squamous cell carcinoma in vitro and in vivo [175]. In a study by Liu et al., the plasmid coding anti‐HER2–CD28‐CD3ζCAR was transfected into NK‐92 cells by electroporation, and the cells specifically killed the ErbB2‐expressing human breast cancer cell lines MDA‐MB‐453 and SKBr3. The adoptive transfer of NK‐92 cells specifically reduced the tumor size and lung metastasis of nude mice transplanted with MDA‐MB‐453 cells and significantly prolonged the survival of these mice [176].

As shown in Table 3, CAR against various tumor‐associated antigens (TAA) has also been evaluated in NK cells. Chang et al. reported a modification by CAR using NKG2D, one of the human NK cell activating receptors, instead of the antigen‐binding site of the antibody. Since NKG2D can bind to eight types of ligands expressed in solid tumors and blood tumors, it can be applied to a broader range of tumor cells. Primary human NK cells were transduced with a retroviral vector, using constructs designed to combine the extracellular domain of NKG2D with CD3ζ and further to express DAP10 simultaneously. This approach showed strong cytotoxic activity against various tumor cell lines and showed no damage to normal cells. It also showed strong tumor growth suppression in a mouse model of osteosarcoma [177].

Advertisement

6. Conclusions

Because NK cells are difficult to culture and it is a challenge to transduce foreign genes into NK cells, research concerning NK cells has been delayed compared to research involving T cells. However, culture and gene transfer technologies for NK cells are now developing. As introduced here, genetically modified NK cells acquired enhanced antitumor functions. These NK cells are very intriguing and are expected to be revolutionary cellular medicines for the treatment of malignancies.

NK cells are heterogeneous populations that exhibit various maturation stages and different KIR expression patterns. Since this heterogeneity has not been completely elucidated, it is not easy to choose the appropriate subset of NK cells for cancer treatment. Although allogeneic NK cell therapy using a KIR mismatch shows a strong antitumor effect against several blood cancers, the mechanisms underlying the activation and maintenance of NK cells in cancer patients are not completely understood. Cancer patients usually undergo a variety of standard treatments before receiving immunotherapy, and it is important to understand the factors that influence NK cell activity in order to select the correct clinical setting for NK cell therapy. In this review, we have explained the combination with antibodies, the genetic modification technique, and the KIR mismatch pattern (which is the basis of patient selection) that has been tested to maximize the use of NK cells as a treatment for cancer. Products of NK cells for clinical use that have been developed worldwide are shown in Table 4.

Gamida Cell Lth.Nant Kwest Inc.Glycostem TherapeuticsGreenCross LabCellFate Therapeutics Inc.GAIA BioMedicine Inc.
H.Q.IsraelUSANetherlandsKoreaUSAJapan
Auto/Allo???Allogeneic (Cell line)AllogeneicAllogeneic (Health donor)AllogeneicAllogeneic (Haploidentical)
MaterialsPBMC/BM/CBNK‐92CB CD34+ (CliniMACS)PBMCPBMC (CMV+ donnor)/iPSPBMC(CliniMACS Prodigy)
Feeder cellIrradiated (2000 rad) PBMC
IrradiationBefore administration
Purity>97% (CD3−/CD56+)>98.1 ± 0.88% (CD3−/CD56+)>98.1 ± 0.88% (CD3−/CD56+)>90.9 ± 2.2% (CD3−/CD56+)>98.0% (CD3-/CD56+)
K562 killingE:T=1:1, 40–50%E:T=5:1 (4 hr), 50%E:T=2:1 (4 hr), 40%E:T=10:1 (4 hr), 70–80%E:T=1:1 (2 hr), >80%
HandlingCPCWash, Irradiation, CPCCPCCPCCPCCompletely closed system
Companion diagnosisHLA/KIR selection
R & D PhaseNon‐clinical(Phase I/II in 2017)Phase IIPhase IPhase IINon‐clinical(Phase I in 2017)Non‐clinical(Phase I in 2017)
Target disease-Merkel cell carcinomaAMLHepatocellular carcinomaAMLNSCLC, Neuroblastoma

Table 4.

Products of NK cells for clinical use.

Advances in the understanding of NK cells may also lead to the development of small‐molecule inhibitors targeting intracellular signals. Because NK cells are difficult to handle, their development is delayed compared to several other immunotherapies, but it is highly likely that they will be established as innovative cell therapy in the near future.

Advertisement

Conflicts of interest

Y. Harada is a member of the Scientific Advisory Boards of GAIA BioMedicine Inc. K. Teraishi is a researcher from Ono Pharmaceutical Co., Ltd. H Ban is a researcher from GAIA BioMedicine Inc. Y. Yonemitsu is a member of the Board of Directors on Science and Medicine at GAIA BioMedicine Inc.

References

  1. 1. Alici E, Konstantinidis KV, Sutlu T et al. Anti‐myeloma activity of endogenous and adoptively transferred activated natural killer cells in experimental multiple myeloma model. Experimental Hematology. 2007;35(12):1839-1846. DOI: 10.1016/j.exphem.2007.08.006
  2. 2. Basse PH, Whiteside TL, Herberman RB. Cancer immunotherapy with interleukin‐2‐activated natural killer cells. Molecular Biotechnology. 2002;21(2):161-170. DOI: 10.1385/MB:21:2:161
  3. 3. Siegler U, Kalberer CP, Nowbakht P, Sendelov S, Meyer‐Monard S, Wodnar‐Filipowicz A. Activated natural killer cells from patients with acute myeloid leukemia are cytotoxic against autologous leukemic blasts in NOD/SCID mice. Leukemia. 2005;19(12):2215-2222. DOI: 10.1038/sj.leu.2403985
  4. 4. Torelli GF, Rozera C, Santodonato L et al. A good manufacturing practice method to ex vivo expand natural killer cells for clinical use. Blood Transfusion. 2015;13(3):464-471. DOI: 10.2450/2015.0231-14
  5. 5. 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. Cancer. 2005;105(8):3051-3057. DOI: 10.1182/blood‐2004‐07‐2974.Supported
  6. 6. Ishikawa E, Tsuboi K, Saijo K, et al. Autologous natural killer cell therapy for human recurrent malignant glioma. Anticancer Research. 2004;24(3 B):1861-1871
  7. 7. 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. Journal of Clinical Oncology. 2010;28(6):955-959. DOI: 10.1200/JCO.2009.24.4590
  8. 8. Carlens S, Gilljam M, Chambers BJ, et al. A new method for in vitro expansion of cytotoxic human CD3–CD56+ natural killer cells. Human Immunology. 2001;62(10):1092-1098. DOI: 10.1016/S0198‐8859(01)00313‐5
  9. 9. Alici E, Sutlu T, Bjo B. et al. Autologous antitumor activity by NK cells expanded from myeloma patients using GMP‐compliant components. Blood. 2008;111(6):3155-3163. DOI: 10.1182/blood‐2007‐09‐110312
  10. 10. Sutlu T, Stellan B, Gilljam M. et al. Clinical‐grade, large‐scale, feeder‐free expansion of highly active human natural killer cells for adoptive immunotherapy using an automated bioreactor. Cytotherapy. 2010;12(8):1044-1055. DOI: 10.3109/14653249.2010.504770
  11. 11. Bachanova V, Linda J. Burns DHM. Allogeneic natural killer cells for refractory. Cancer Immunology, Immunotherapy. 2010;59(11):1739-1744. DOI: 10.1007/s00262‐010‐0896‐z.ALLOGENEIC
  12. 12. Koehl U, Brehm C, Huenecke S. et al. Clinical grade purification and expansion of NK cell products for an optimized manufacturing protocol. Frontiers in Oncology. 2013;3. DOI: 10.3389/fonc.2013.00118
  13. 13. Saito S, Harada Y, Morodomi Y. et al. Ex vivo generation of highly purified and activated natural killer cells from human peripheral blood. Human Gene Therapy Methods. 2013;24(4):241-252. DOI: 10.1089/hgtb.2012.183
  14. 14. Anfossi N, André P, Guia S, et al. Human NK cell education by inhibitory receptors for MHC class I. Immunity. 2006;25(2):331-342. DOI: 10.1016/j.immuni.2006.06.013
  15. 15. Lucas M, Schachterle W, Oberle K, Aichele P, Andreas D. Natural killer cell‐mediated control of infections requires production of interleukin 15 by Type I IFN‐triggered dendritic cells. Immunity. 2007;26:503-517. DOI: 10.1016/j.biotechadv.2011.08.021.Secreted
  16. 16. Mortier E, Advincula R, Kim L, et al. Macrophage‐ and dendritic‐cell‐derived interleukin‐15 receptor alpha supports homeostasis of distinct CD8+ T cell subsets. Immunity. 2009;31(5):811-822. DOI: 10.1016/j.immuni.2009.09.017
  17. 17. Brehm C, Huenecke S, Quaiser A, et al. IL‐2 stimulated but not unstimulated NK cells induce selective disappearance of peripheral blood cells: Concomitant results to a phase I/II study. PLoS One. 2011;6(11). DOI: 10.1371/journal.pone.0027351
  18. 18. Guia S, Cognet C, de Beaucoudrey L, et al. A role for interleukin‐12/23 in the maturation of human natural killer and CD56+ T cells in vivo. Blood. 2008;111(10):5008-5016. DOI: 10.1182/blood‐2007‐11‐122259
  19. 19. Chaix J, Tessmer MS, Hoebe K, et al. Priming of natural killer cells by Interleukin‐18. J Immunol. 2008;181(3):1627-1631
  20. 20. Lapteva N, Durett AG, Sun J, et al. Large‐scale ex vivo expansion and characterization of natural killer cells for clinical applications. Cytotherapy. 2012;14(9):1131-1143. DOI: 10.3109/14653249.2012.700767
  21. 21. Cooper MA, Bush JE, Fehniger TA. et al. In vivo evidence for a dependence on interleukin 15 for survival of natural killer cells. Blood. 2002;100(10):3633-3638. DOI: 10.1182/blood‐2001‐12‐0293
  22. 22. Dahlberg CIM, Sarhan D, Chrobok M, Duru AD, Alici E. Natural killer cell‐based therapies targeting cancer: Possible strategies to gain and sustain anti‐tumor activity. Frontiers in Immunology. 2015;6(NOV). DOI: 10.3389/fimmu.2015.00605
  23. 23. Rham de C, Ferrari‐Lacraz S, Jendly S, Schneiter G, et al. The proinflammatory cytokines IL‐2, IL‐15 and IL‐21 modulate the repertoire of mature human natural killer cell receptors. Arthritis Research & Therapy. 2007;9:R125. DOI: 10.1186/ar2336
  24. 24. Berg M, Lundqvist A, McCoy P, et al. Clinical‐grade ex vivo‐expanded human natural killer cells up‐regulate activating receptors and death receptor ligands and have enhanced cytolytic activity against tumor cells. Cytotherapy. 2009;11(3):341-355. DOI: 10.1080/14653240902807034
  25. 25. Ljunggren HG, Kärre K. In search of the “missing self”: MHC molecules and NK cell recognition. Immunology Today. 1990;11(C):237-244. DOI: 10.1016/0167‐5699(90)90097‐S
  26. 26. Hudson LE, Allen RL. Leukocyte Ig‐like receptors—A model for MHC class I disease associations. Frontiers in Immunology. 2016;7(JUL):1-8. DOI: 10.3389/fimmu.2016.00281
  27. 27. Chester C, Fritsch K, Kohrt HE. Natural killer cell immunomodulation: Targeting activating, inhibitory, and co‐stimulatory receptor signaling for cancer immunotherapy. Frontiers in Immunology. 2015;6(DEC):1-9. DOI: 10.3389/fimmu.2015.00601
  28. 28. Vivier E, Raulet D, Moretta A, Caligiuri M. Innate or adaptive immunity? The example of natural killer cells. Science (80). 2011;331(6013):44-49. DOI: 10.1126/science.1198687.Innate
  29. 29. Vivier E, Nunes JA, Vely F. Natural killer cell signaling pathways. Science. 2004;306(5701):1517-1519. DOI: 306/5701/1517 [pii]\n10.1126/science.1103478
  30. 30. Holmes TD, El‐Sherbiny YM, Davison A, Clough SL, Blair GE, Cook GP. A human NK cell activation/inhibition threshold allows small changes in the target cell surface phenotype to dramatically alter susceptibility to NK cells. The Journal of Immunology. 2011;186(3):1538-1545. DOI: 10.4049/jimmunol.1000951
  31. 31. Varbanova V, Naumova E, Mihaylova A. Killer‐cell immunoglobulin‐like receptor genes and ligands and their role in hematologic malignancies. Cancer Immunology, Immunotherapy. 2016;65(4):427-440. DOI: 10.1007/s00262‐016‐1806‐9
  32. 32. Wilson MJ, Torkar M, Haude A et al. Plasticity in the organization and sequences of human KIR/ILT gene families. Proceedings of the National Academy of Sciences USA. 2000;97(9):4778-4783. DOI: 10.1073/pnas.080588597
  33. 33. Hsu KC, Chida S, Geraghty DE, Dupont B. The killer cell immunoglobulin‐like receptor (KIR) genomic region: Gene‐order, haplotypes and allelic polymorphism. Immunological Reviews. 2002;190:40-52. DOI: imr19004 [pii]
  34. 34. 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;113(3):726-732. DOI: 10.1182/blood‐2008‐07‐171926
  35. 35. Symons HJ, Leffell MS, Rossiter ND, Zahurak M, Jones RJ, Fuchs EJ. Transplantation. Blood. 2011;16(4):533-542. DOI: 10.1016/j.bbmt.2009.11.022.Improved
  36. 36. Leung W, Handgretinger R, Iyengar R, Turner V, Holladay MS, Hale Ga. Inhibitory KIR‐HLA receptor‐ligand mismatch in autologous haematopoietic stem cell transplantation for solid tumour and lymphoma. British Journal of Cancer. 2007;97(4):539-542. DOI: 10.1038/sj.bjc.6603913
  37. 37. Colonna M, Borsellino G, Falco M, Ferrara GB, Strominger JL. HLA‐C is the inhibitory ligand that determines dominant resistance to lysis by NK1‐ and NK2‐specific natural killer cells. Proceedings of the National Academy of Sciences USA. 1993;90(24):12000-12004. DOI: 10.1073/pnas.90.24.12000
  38. 38. Moesta AK, Norman PJ, Yawata M, Yawata N, Gleimer M, Parham P. Synergistic polymorphism at two positions distal to the ligand‐binding site makes KIR2DL2 a stronger receptor for HLA‐C than KIR2DL3. The Journal of Immunology. 2008;180(6):3969-3979. DOI: 10.4049/JIMMUNOL.180.6.3969
  39. 39. Yawata M, Yawata N, Draghi M, Little A‐M, Partheniou F, Parham P. Roles for HLA and KIR polymorphisms in natural killer cell repertoire selection and modulation of effector function. The Journal of Experimental Medicine. 2006;203(3):633-645. DOI: 10.1084/jem.20051884
  40. 40. Dohring C, Scheidegger D, Samaridis J, Cella M, Colonna M. A human killer inhibitory receptor specific for HLA‐A1, 2. The Journal of Immunology. 1996;156(9):3098-3101. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=8617928
  41. 41. Hilton HG, Guethlein LA, Goyos A, et al. Polymorphic HLA‐C receptors balance the functional characteristics of KIR haplotypes. The Journal of Immunology. 2015;195:3160-3170. DOI: 10.4049/jimmunol.1501358
  42. 42. Smyth MJ, Cretney E, Kelly JM, et al. Activation of NK cell cytotoxicity. Molecular Immunology. 2005;42(4 SPEC. ISS.):501-510. DOI: 10.1016/j.molimm.2004.07.034
  43. 43. Ikner A, Ashkenazi A. TWEAK induces apoptosis through a death‐signaling complex comprising receptor‐interacting protein 1 (RIP1), Fas‐associated death domain (FADD), and caspase‐8. The Journal of Biological Chemistry. 2011;286(24):21546-21554. DOI: 10.1074/jbc.M110.203745
  44. 44. Degli‐Esposti M. To die or not to die—the quest of the TRAIL receptors. Journal of Leukocyte Biology. 1999;65(5):535-542. http://www.ncbi.nlm.nih.gov/pubmed/10331480
  45. 45. Smyth MJ, Takeda K, Hayakawa Y, Peschon JJ, van den Brink MR., Yagita H. Nature’s TRAIL—On a Path to Cancer Immunotherapy. Immunity. 2003;18(1):1-6. DOI: 10.1016/S1074‐7613(02)00502‐2
  46. 46. Wajant H, Moosmayer D, Wüest T, et al. Differential activation of TRAIL‐R1 and ‐2 by soluble and membrane TRAIL allows selective surface antigen‐directed activation of TRAIL‐R2 by a soluble TRAIL derivative. Oncogene. 2001;20(30):4101-4106. DOI: 10.1038/sj.onc.1204558
  47. 47. Guicciardi ME, Gores GJ. Life and death by death receptors. The FASEB Journal. 2009;23(6):1625-1637. DOI: 10.1096/fj.08‐111005
  48. 48. Griffith BTS, Wiley SR, Kubin MZ, Sedger LM, Maliszewski CR, Fanger N. A necrosis factor‐ related cytokine, TRAIL. Cell. 1999;189(8)
  49. 49. Fanger NA, Maliszewski CR, Schooley K, Griffith TS. Human dendritic cells mediate cellular apoptosis via tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL). The Journal of Experimental Medicine. 1999;190(8):1155-1164. DOI: 10.1084/jem.190.8.1155
  50. 50. Smyth MJ, Cretney E, Takeda K. et al. Tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL) contributes to interferon gamma‐dependent natural killer cell protection from tumor metastasis. The Journal of Experimental Medicine. 2001;193(6):661-670. DOI: 10.1084/jem.193.6.661
  51. 51. Screpanti V, Wallin RPA, Grandien A, Ljunggren HG. Impact of FASL‐induced apoptosis in the elimination of tumor cells by NK cells. Molecular Immunology. 2005;42(4 SPEC. ISS.):495-499. DOI: 10.1016/j.molimm.2004.07.033
  52. 52. Wallin RPA, Screpanti V, Michaëlsson J, Grandien A, Ljunggren HG. Regulation of perforin‐independent NK cell‐mediated cytotoxicity. European Journal of Immunology. 2003;33(10):2727-2735. DOI: 10.1002/eji.200324070
  53. 53. Reichmann E. The biological role of the Fas/FasL system during tumor formation and progression. Seminars in Cancer Biology. 2002;12:309-315
  54. 54. Voskoboinik I, Whisstock JC, Trapani Ja. Perforin and granzymes: function, dysfunction and human pathology. Nature Reviews Immunology. 2015;15(6):388-400. DOI: 10.1038/nri3839
  55. 55. Wulfing C, Purtic B, Klem J, Schatzle JD. Stepwise cytoskeletal polarization as a series of checkpoints in innate but not adaptive cytolytic killing. Proceedings of the National Academy of Sciences USA. 2003;100(13):7767-7772. DOI: 10.1073/pnas.1336920100
  56. 56. DiPersio JF, Uy GL, Yasothan U. et al. Plerixafor. Nature Reviews Drug Discovery. 2009;8(2):105-106. DOI: 10.1038/nrd2819
  57. 57. DiPersio JF, Micallef IN, Stiff PJ, et al. Phase III prospective randomized double‐blind placebo‐controlled trial of plerixafor plus granulocyte colony‐stimulating factor compared with placebo plus granulocyte colony‐stimulating factor for autologous stem‐cell mobilization and transplantation for patients with non‐Hodgkin’s lymphoma. Journal of Clinical Oncology. 2009;27(28):4767-73. DOI: 10.1200/JCO.2008.20.7209
  58. 58. Nademanee AP, DiPersio JF, Maziarz RT, et al. Plerixafor plus granulocyte colony‐stimulating factor versus placebo plus granulocyte colony‐stimulating factor for mobilization of CD34(+) hematopoietic stem cells in patients with multiple myeloma and low peripheral blood CD34(+) cell count: results of a subset analysis of a randomized trial. Biology of Blood and Marrow Transplantation. 2012;18(10):1564-1572. DOI: 10.1016/j.bbmt.2012.05.017
  59. 59. Gabrilovich DI, Nagaraj S. Myeloid‐derived suppressor cells as regulators of the immune system. Nature Reviews Immunology. 2009;9(3):162-174. DOI: 10.1038/nri2506
  60. 60. Luyckx A, Schouppe E, Rutgeerts O, et al. G‐CSF stem cell mobilization in human donors induces polymorphonuclear and mononuclear myeloid‐derived suppressor cells. Clinical Immunology. 2012;143(1):83-87. DOI: 10.1016/j.clim.2012.01.011
  61. 61. Gluckman E, Broxmeyer HA, Auerbach AD. et al. Hematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical‐cord blood from an HLA‐identical sibling. The New England Journal of Medicine. 1989;321(17):1174-1178. DOI: 10.1056/NEJM198910263211707
  62. 62. Eapen M, Rubinstein P, Zhang MJ. et al. Outcomes of transplantation of unrelated donor umbilical cord blood and bone marrow in children with acute leukaemia: a comparison study. Lancet. 2007;369(9577):1947-1954. DOI: 10.1016/S0140‐6736(07)60915‐5
  63. 63. Atsuta Y, Suzuki R, Nagamura‐Inoue T, et al. Disease‐specific analyses of unrelated cord blood transplantation compared with unrelated bone marrow transplantation in adult patients with acute leukemia. Blood. 2009;113(8):1631-1638. DOI: 10.1182/blood‐2008‐03‐147041
  64. 64. Takahashi S, Ooi J, Tomonari A, et al. Comparative single‐institute analysis of cord blood transplantation from unrelated donors with bone marrow or peripheral blood stem‐cell transplants from related donors in adult patients with hematologic malignancies after myeloablative conditioning regimen. Blood. 2007;109(3):1322-1330. DOI: 10.1182/blood‐2006‐04‐020172
  65. 65. Woll PS, Martin CH, Miller JS. et al. Human embryonic stem cell‐derived NK cells acquire functional receptors and cytolytic activity. The Journal of Immunology. 2005;175(8):5095-5103
  66. 66. Woll PS, Grzywacz B, Tian X. et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood. 2009;113(24):6094-6101. DOI: 10.1182/blood‐2008‐06‐165225
  67. 67. Rezvani K, Rouce RH. The application of natural killer cell immunotherapy for the treatment of cancer. Frontiers in Immunology. 2015;6:578. DOI: 10.3389/fimmu.2015.00578
  68. 68. Boissel L, Betancur‐Boissel M, Lu W. et al. Retargeting NK‐92 cells by means of CD19‐ and CD20‐specific chimeric antigen receptors compares favorably with antibody‐dependent cellular cytotoxicity. Oncoimmunology. 2013;2(10): e26527. DOI: 10.4161/onci.26527
  69. 69. Hermanson DL, Kaufman DS. Utilizing chimeric antigen receptors to direct natural killer cell activity. Frontiers in Immunology. 2015;6:195. DOI: 10.3389/fimmu.2015.00195
  70. 70. Gong JH, Maki G, Klingemann HG. Characterization of a human cell line (NK‐92) with phenotypical and functional characteristics of activated natural killer cells. Leukemia. 1994;8(4):652-658
  71. 71. Yan Y, Steinherz P, Klingemann HG. et al. Antileukemia activity of a natural killer cell line against human leukemias. Clinical Cancer Research. 1998;4(11):2859-2868
  72. 72. Jochems C, Hodge JW, Fantini M. et al. An NK cell line (haNK) expressing high levels of granzyme and engineered to express the high affinity CD16 allele. Oncotarget. 2016;7(52):86359-86373. DOI: 10.18632/oncotarget.13411
  73. 73. Klingemann HG, Martinson J. Ex vivo expansion of natural killer cells for clinical applications. Cytotherapy. 2004;6(1):15-22. DOI: 10.1080/14653240310004548
  74. 74. Iliopoulou EG, Kountourakis P, Karamouzis MV. et al. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non‐small cell lung cancer. Cancer Immunology, Immunotherapy. 2010;59(12):1781-1789. DOI: 10.1007/s00262‐010‐0904‐3
  75. 75. Yoon SR, Lee YS, Yang SH. et al. Generation of donor natural killer cells from CD34(+) progenitor cells and subsequent infusion after HLA‐mismatched allogeneic hematopoietic cell transplantation: a feasibility study. Bone Marrow Transplant Centre. 2010;45(6):1038-1046. DOI: 10.1038/bmt.2009.304
  76. 76. Ozaki K, Kikly K, Michalovich D. et al. Cloning of a type I cytokine receptor most related to the IL‐2 receptor beta chain. Proceedings of the National Academy of Sciences U S A. 2000;97(21):11439-11444. DOI: 10.1073/pnas.200360997
  77. 77. Parrish‐Novak J, Dillon SR, Nelson A. et al. Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature. 2000;408(6808):57-63. DOI: 10.1038/35040504
  78. 78. Leonard WJ, Spolski R. Interleukin‐21: a modulator of lymphoid proliferation, apoptosis and differentiation. Nature Reviews Immunology. 2005;5(9):688-698. DOI: 10.1038/nri1688
  79. 79. Shruti Bhatt, Julie Matthews, Salma Parvin. et al. Direct and immune‐mediated cytotoxicity of interleukin‐21 contributes to antitumor effects in mantle cell lymphoma. Blood. 2015;126(13):1555-1564. DOI: 10.1182/blood‐2015‐01‐624585
  80. 80. Miller JS, Oelkers S, Verfaillie C. et al. Role of monocytes in the expansion of human activated natural killer cells. Blood. 1992;80(9):2221-2229
  81. 81. Boissel L, Tuncer HH, Betancur M. et al. Umbilical cord mesenchymal stem cells increase expansion of cord blood natural killer cells. Biology of Blood and Marrow Transplantation. 2008;14(9):1031-1038. DOI: 10.1016/j.bbmt.2008.06.016
  82. 82. Shah N, Martin‐Antonio B, Yang H. et al. Antigen presenting cell‐mediated expansion of human umbilical cord blood yields log‐scale expansion of natural killer cells with anti‐myeloma activity. PLoS One. 2013;8(10):e76781. DOI: 10.1371/journal.pone.0076781
  83. 83. Granzin M, Soltenborn S, Müller S. et al. Fully automated expansion and activation of clinical‐grade natural killer cells for adoptive immunotherapy. Cytotherapy. 2015;17(5):621-632. DOI: 10.1016/j.jcyt.2015.03.611
  84. 84. Geraghty RJ, Capes‐Davis A, Davis JM. et al. Guidelines for the use of cell lines in biomedical research. British Journal of Cancer. 2014;111(6):1021-1046. DOI: 10.1038/bjc.2014.166
  85. 85. Spanholtz J, Preijers F, Tordoir M. et al. Clinical‐grade generation of active NK cells from cord blood hematopoietic progenitor cells for immunotherapy using a closed‐system culture process. PLoS One. 2011;6(6):e20740. DOI: 10.1016/j.msard.2012.01.001
  86. 86. Knorr DA, Ni Z, Hermanson D. et al. Clinical‐scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Translational Medicine. 2013;2(4):274-283. DOI: 10.5966/sctm.2012‐0084
  87. 87. Krause SW, Gastpar R, Andreesen R. et al. Treatment of colon and lung cancer patients with ex vivo heat shock protein 70‐peptide‐activated, autologous natural killer cells: A clinical phase 1 trial. Clinical Cancer Research. 2004;10(11):3699-3707. DOI: 10.1158/1078‐0432.CCR‐03‐0683
  88. 88. Parkhurst MR, Riley JP, Dudley ME. et al. Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clinical Cancer Research. 2011;17(19):6287-6297. DOI: 10.1158/1078‐0432.CCR‐11‐1347
  89. 89. Sakamoto N, Ishikawa T, Kokura S. et al. Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. Journal of Translational Medicine. 2015;13:277. DOI: 10.1186/s12967‐015‐0632‐8
  90. 90. Koehl U, Sörensen J, Esser R. et al. IL‐2 activated NK cell immunotherapy of three children after haploidentical stem cell transplantation. Blood Cells, Molecules and Diseases. 2004;33(3):261-266. DOI: 10.1016/j.bcmd.2004.08.013
  91. 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;125(5):784-792. DOI: 10.1182/blood‐2014‐07‐592881
  92. 92. Lanier LL, Ruitenberg JJ, Phillips JH. Functional and biochemical‐analysis of Cd16 antigen on natural‐killer cells and granulocytes. The Journal of Immunology. 1988;141(10):3478-3485
  93. 93. Morel PA, Ernst LK, Metes D. Functional CD32 molecules on human NK cells. Leukemia & Lymphoma. 1999;35(1-2):47-56. DOI: 10.3109/10428199909145704
  94. 94. Bruhns P, Iannascoli B, England P. et al. Specificity and affinity of human Fc gamma receptors and their polymorphic variants for human IgG subclasses. Blood. 2009;113(16):3716-3725. DOI: 10.1182/Blood‐2008‐09‐179754
  95. 95. Hogarth PM, Pietersz GA. Fc receptor‐targeted therapies for the treatment of inflammation, cancer and beyond. Nature Reviews Drug Discovery. 2012;11(4):311-331. DOI: 10.1038/nrd2909
  96. 96. Breunis WB, Van Mirre E, Geissler J. et al. Copy number variation at the FCGR locus includes FCGR3A, FCGR2C and FCGR3B but not FCGR2A and FCGR2B. Human Mutation. 2009;30(5):E640‐E650. DOI: 10.1002/humu.20997
  97. 97. Wang W, Erbe AK, Hank JA. et al. NK cell‐mediated antibody‐dependent cellular cytotoxicity in cancer immunotherapy. Frontiers in Immunology. 2015;6:368. DOI: 10.3389/fimmu.2015.00368
  98. 98. Cartron G, Dacheux L, Salles G. et al. Therapeutic activity of humanized anti‐CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood. 2002;99(3):754-758
  99. 99. Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. Journal of Clinical Oncology. 2003;21(21):3940-3947. DOI: 10.1200/JCO.2003.05.013
  100. 100. Binyamin L, Alpaugh RK, Hughes TL. et al. Blocking NK cell inhibitory self‐recognition promotes antibody‐dependent cellular cytotoxicity in a model of anti‐lymphoma therapy. The Journal of Immunology. 2008;180(9):6392-6401. DOI: 10.4049/jimmunol.180.9.6392
  101. 101. Carlsten M, Levy E, Karambelkar A. et al. Efficient mRNA‐Based genetic engineering of human NK cells with high‐affinity CD16 and CCR7 augments rituximab‐induced ADCC against lymphoma and targets NK Cell migration toward the lymph node‐associated chemokine CCL19. Frontiers in Immunology. 2016;7:105. DOI: 10.3389/fimmu.2016.00105
  102. 102. Romee R, Foley B, Lenvik T. et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease‐17 (ADAM17). Blood. 2013;121(18):3599-3608. DOI: 10.1182/blood‐2012‐04‐425397
  103. 103. Wiernik A, Foley B, Zhang B. et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16 x 33 bispecific killer cell engager and ADAM17 inhibition. Clinical Cancer Research. 2013;19(14):3844-3855. DOI: 10.1158/1078‐0432.CCR‐13‐0505
  104. 104. Jing Y, Ni Z, Wu J. et al. Identification of an ADAM17 cleavage region in human CD16 (FcγRIII) and the engineering of a non‐cleavable version of the receptor in NK cells. PLoS One. 2015;10(3):e0121788. DOI: 10.1371/journal.pone.0121788
  105. 105. Ruggeri L, Mancusi A, Burchielli E. et al. NK cell alloreactivity and allogeneic hematopoietic stem cell transplantation. Blood Cells, Molecules and Diseases. 2008;40(1):84-90. DOI: 10.1016/j.bcmd.2007.06.029
  106. 106. Morishima Y, Yabe T, Matsuo K. et al. Effects of HLA allele and killer immunoglobulin‐like receptor ligand matching on clinical outcome in leukemia patients undergoing transplantation with T‐cell‐replete marrow from an unrelated donor. Biology of Blood and Marrow Transplantation. 2007;13(3):315-328. DOI: 10.1016/j.bbmt.2006.10.027
  107. 107. Farag SS, Bacigalupo A, Eapen M. et al. 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. Biology of Blood and Marrow Transplantation. 2006;12(8):876-884. DOI: 10.1016/j.bbmt.2006.05.007
  108. 108. Yabe T, Matsuo K, Hirayasu K, et al. Donor killer immunoglobulin‐like receptor (KIR) genotype‐patient cognate KIR ligand combination and antithymocyte globulin preadministration are critical factors in outcome of HLA‐C‐KIR ligand‐mismatched T cell‐replete unrelated bone marrow transplantation. Biology of Blood and Marrow Transplantation. 2008;14(1):75-87. DOI: 10.1016/j.bbmt.2007.09.012
  109. 109. Yazaki M, Takahashi T, Mizutani K. et al. Human leucocyte antigen‐Cw‐specific cytotoxic T lymphocytes generated from naive cord blood used for cord blood stem cell transplantation. Biology of Blood and Marrow Transplantation. 2002;117(4):893-898
  110. 110. Sugimoto K, Murata M, Terakura S. et al. CTL clones isolated from an HLA‐Cw‐mismatched bone marrow transplant recipient with acute graft‐versus‐host disease. The Journal of Immunology. 2009;183(9):5991-5998. DOI: 10.4049/jimmunol.0804310
  111. 111. Leung W, Iyengar R, Turner V. et al. Determinants of antileukemia effects of allogeneic NK cells. The Journal of Immunology. 2004;172(1):644-650
  112. 112. 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;105(12):4878-4884. DOI: 10.1182/blood‐2004‐12‐4825
  113. 113. Hsu KC, Gooley T, Malkki M. et al. KIR ligands and prediction of relapse after unrelated donor hematopoietic cell transplantation for hematologic malignancy. Biology of Blood and Marrow Transplantation. 2006;12(8):828-836. DOI: 10.1016/j.bbmt.2006.04.008
  114. 114. Miller JS, Cooley S, Parham P. et al. Missing KIR ligands are associated with less relapse and increased graft‐versus‐host disease (GVHD) following unrelated donor allogeneic HCT. Blood. 2007;109(11):5058-5061. DOI: 10.1182/blood‐2007‐01‐065383
  115. 115. 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;110(1):433-440. DOI: 10.1182/blood‐2006‐07‐038687
  116. 116. 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;118(12):3273-3279. DOI: 10.1182/blood‐2011‐01‐329508
  117. 117. Farnault L, Sanchez C, Baier C. et al. Hematological malignancies escape from NK cell innate immune surveillance: Mechanisms and therapeutic implications. Clinical and Developmental Immunology. 2012;2012:421702. DOI: 10.1155/2012/421702
  118. 118. Fauriat C, Andersson S, Björklund AT. et al. Estimation of the size of the alloreactive NK cell repertoire: studies in individuals homozygous for the group A KIR haplotype. The Journal of Immunology. 2008;181(9):6010-6019
  119. 119. 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;116(14):2411-2419. DOI: 10.1182/blood‐2010‐05‐283051
  120. 120. Leung W, Use of NK cell activity in cure by transplant. British Journal of Haematology. 2011;155(1):14-29. DOI: 10.1111/j.1365‐2141.2011
  121. 121. 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;113(13):3119-3329. DOI: 10.1182/blood‐2008‐06‐164103
  122. 122. Venstrom JM, Pittari G, Gooley TA. et al. HLA‐C‐dependent prevention of leukemia relapse by donor activating KIR2DS1. The New England Journal of Medicine. 2012;367(9):805-816. DOI: 10.1056/NEJMoa1200503
  123. 123. Venstrom JM, Gooley TA, Spellman S. et al. Donor activating KIR3DS1 is associated with decreased acute GVHD in unrelated allogeneic hematopoietic stem cell transplantation. Blood. 2010;115(15):3162-3165. DOI: 10.1182/blood‐2009‐08‐236943
  124. 124. Ludajic K, Balavarca Y, Bickeböller H. et al. KIR genes and KIR ligands affect occurrence of acute GVHD after unrelated, 12/12 HLA matched, hematopoietic stem cell transplantation. Bone Marrow Transplant Centre. 2009;44(2):97-103. DOI: 10.1038/bmt.2008.432
  125. 125. Littera R, Orrù N, Vacca A. et al. The role of killer immunoglobulin‐like receptor haplotypes on the outcome of unrelated donor haematopoietic SCT for thalassaemia. Bone Marrow Transplant Centre. 2010;45(11):1618-1624. DOI: 10.1038/bmt.2010.24
  126. 126. Callahan MK, Postow MA, Wolchok JD. CTLA‐4 and PD‐1 Pathway Blockade: Combinations in the Clinic. Frontiers in Oncology. 2015;4:385. DOI: 10.3389/fonc.2014.00385
  127. 127. Valsecchi ME. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. The New England Journal of Medicine. 2015;373(13):1270. DOI: 10.1056/NEJMc1509660#SA1
  128. 128. Benson DM Jr, Bakan CE, Mishra A. et al. The PD‐1/PD‐L1 axis modulates the natural killer cell versus multiple myeloma effect: A therapeutic target for CT‐011, a novel monoclonal anti‐PD‐1 antibody. Blood. 2010;116(13):2286-2294. DOI: 10.1182/blood‐2010‐02‐271874
  129. 129. Campbell KS, Hasegawa J. Natural killer cell biology: an update and future directions. Journal of Allergy and Clinical Immunology. 2013;132(3):536-544. DOI: 10.1016/j.jaci.2013.07.006
  130. 130. Garrido F, Cabrera T, Accolla RS. et al. Functional and medical implications. vol. I. In: Charron D, EDK (eds). 1997. pp 445-452
  131. 131. Marincola FM, Jaffee EM, Hicklin DJ. et al. Escape of human solid tumors from T‐cell recognition: molecular mechanisms and functional significance. Advances in Immunology. 2000;74:181-273
  132. 132. Campoli M, Chang CC, Ferrone S. HLA class I antigen loss, tumor immune escape and immune selection. Vaccine. 2002;20 Suppl 4: A40‐A45
  133. 133. Chang CC, Campoli M, Ferrone S. Classical and nonclassical HLA class I antigen and NK Cell‐activating ligand changes in malignant cells: current challenges and future directions. Advances in Cancer Research. 2005;93:189-234. DOI: 10.1016/S0065‐230X(05)93006‐6
  134. 134. Aptsiauri N, Cabrera T, Mendez R. et al. Role of altered expression of HLA class I molecules in cancer progression. Advances in Experimental Medicine and Biology. 2007;601:123-31
  135. 135. Blades RA, Keating PJ, McWilliam LJ. et al. Loss of HLA class I expression in prostate cancer: implications for immunotherapy. Urology. 1995;46(5):681-686; discussion 686-687. DOI: 10.1016/S0090‐4295(99)80301‐X
  136. 136. Cabrera T, Angustias Fernandez M. et al. High frequency of altered HLA class I phenotypes in invasive breast carcinomas. Human Immunology. 1996 50(2):127-134
  137. 137. Cabrera T, Collado A, Fernandez MA. et al. High frequency of altered HLA class I phenotypes in invasive colorectal carcinomas. Tissue Antigens. 1998;52(2):114-123
  138. 138. Cabrera T, Salinero J, Fernandez MA. et al. High frequency of altered HLA class I phenotypes in laryngeal carcinomas. Human Immunology. 2000;61(5):499-506
  139. 139. Koopman LA, Corver WE, van der Slik AR, et al. Multiple genetic alterations cause frequent and heterogeneous human histocompatibility leukocyte antigen class I loss in cervical cancer. The Journal of Experimental Medicine. 2000;191(6):961-976
  140. 140. Kageshita T, Ishihara T, Campoli M, et al. Selective monomorphic and polymorphic HLA class I antigenic determinant loss in surgically removed melanoma lesions. Tissue Antigens. 2005;65(5):419-428
  141. 141. Feenstra M, Verdaasdonk M, van der Zwan AW, et al. Microsatellite analysis of microdissected tumor cells and 6p high density microsatellite analysis in head and neck squamous cell carcinomas with down‐regulated human leukocyte antigen class I expression. Laboratory Investigation. 2000;80(3):405-414
  142. 142. Maleno I, López‐Nevot MA, Cabrera T, et al. Multiple mechanisms generate HLA class I altered phenotypes in laryngeal carcinomas: High frequency of HLA haplotype loss associated with loss of heterozygosity in chromosome region 6p21. Cancer Immunology and Immunotherapy. 2002;51(7):389-396. DOI: 10.1007/s00262‐002‐0296‐0
  143. 143. Maleno I, Cabrera CM, Cabrera T, et al. Distribution of HLA class I altered phenotypes in colorectal carcinomas: High frequency of HLA haplotype loss associated with loss of heterozygosity in chromosome region 6p21. Immunogenetics. 2004;56(4):244-53. DOI: 10.1007/s00251‐004‐0692‐z
  144. 144. Maleno I, Romero JM, Cabrera T, et al. LOH at 6p21.3 region and HLA class I altered phenotypes in bladder carcinomas. Immunogenetics. 2006;58(7):503-510. DOI: 10.1007/s00251‐006‐0111‐8
  145. 145. Carlsten M, Childs RW. Genetic manipulation of NK cells for cancer immunotherapy: techniques and clinical implications. Frontiers in Immunology. 2015;6:266. DOI: 10.3389/fimmu.2015.00266
  146. 146. Sutlu T, Nyström S, Gilljam M. et al. Inhibition of intracellular antiviral defense mechanisms augments lentiviral transduction of human natural killer cells: Implications for gene therapy. Human Gene Therapy. 2012;23(10):1090-1100. DOI: 10.1089/hum.2012.080
  147. 147. Guven H, Konstantinidis KV, Alici E. et al. Efficient gene transfer into primary human natural killer cells by retroviral transduction. Experimental Hematology. 2005;33(11):1320-1328. DOI: 10.1016/j.exphem.2005.07.006
  148. 148. Schirrmann T, Pecher G. Human natural killer cell line modified with a chimeric immunoglobulin T‐cell receptor gene leads to tumor growth inhibition in vivo. Cancer Gene Therapy. 2002r;9(4):390-398. DOI: 10.1038/sj.cgt.7700453
  149. 149. Grund EM, Muise‐Helmericks RC. Cost efficient and effective gene transfer into the human natural killer cell line, NK92. Journal of Immunological Methods. 2005;296(1-2):31-36. DOI: 10.1016/j.jim.2004.10.008
  150. 150. Schirrmann T, Pecher G. Specific targeting of CD33(+) leukemia cells by a natural killer cell line modified with a chimeric receptor. Leukemia Research. 2005;29(3):301-316. DOI: 10.1016/j.leukres.2004.07.005
  151. 151. Boissel L, Betancur M, Wels WS. et al. Transfection with mRNA for CD19 specific chimeric antigen receptor restores NK cell mediated killing of CLL cells. Leukemia Research. 2009;33(9):1255-1259. DOI: 10.1016/j.leukres.2008.11.024
  152. 152. Li L, Liu LN, Feller S. et al. Expression of chimeric antigen receptors in natural killer cells with a regulatory‐compliant non‐viral method. Cancer Gene Therapy. 2010;17(3):147-154. DOI: 10.1038/cgt.2009.61
  153. 153. Shimasaki N, Fujisaki H, Cho D, et al. A clinically adaptable method to enhance the cytotoxicity of natural killer cells against B‐cell malignancies. Cytotherapy. 2012;14(7):830-840. DOI: 10.3109/14653249.2012.671519
  154. 154. Maasho K, Marusina A, Reynolds NM. et al. Efficient gene transfer into the human natural killer cell line, NKL, using the amaxa nucleofection system. Journal of Immunological Methods. 2004;284(1-2):133-140. DOI: 10.1016/j.jim.2003.10.010
  155. 155. Schoenberg K, Trompeter HI, Uhrberg M. Delivery of DNA into natural killer cells for immunotherapy. Methods in Molecular Biology. 2008;423:165-172. DOI: 10.1007/978‐1‐59745‐194‐9_11
  156. 156. Astoul P, Viallat JR, Laurent JC, et al. Intrapleural recombinant IL‐2 in passive immunotherapy for malignant pleural effusion. Chest. 1993;103(1):209-213
  157. 157. Goey SH, Eggermont AM, Punt CJ, et al. Intrapleural administration of interleukin 2 in pleural mesothelioma: A phase I–II study. British Journal of Cancer. 1995;72(5):1283-1288
  158. 158. Nagashima S, Mailliard R, Kashii Y. et al. Stable transduction of the interleukin‐2 gene into human natural killer cell lines and their phenotypic and functional characterization in vitro and in vivo. Blood. 1998;91(10):3850-6381
  159. 159. Imamura M, Shook D, Kamiya T. et al. Autonomous growth and increased cytotoxicity of natural killer cells expressing membrane‐bound interleukin‐15. Blood. 2014;124(7):1081-1088. DOI: 10.1182/blood‐2014‐02‐556837
  160. 160. Sahm C, Schönfeld K, Wels WS. Expression of IL‐15 in NK cells results in rapid enrichment and selective cytotoxicity of gene‐modified effectors that carry a tumor‐specific antigen receptor. Cancer Immunology, Immunotherapy. 2012;61(9):1451-1461. DOI: 10.1007/s00262‐012‐1212‐x
  161. 161. Jiang W, Zhang C, Tian Z, et al. hIL‐15 gene‐modified human natural killer cells (NKL‐IL15) augments the anti‐human hepatocellular carcinoma effect in vivo. Immunobiology. 2014;219(7):547-553. DOI: 10.1016/j.imbio.2014.03.007
  162. 162. Inngjerdingen M, Damaj B, Maghazachi AA. Expression and regulation of chemokine receptors in human natural killer cells. Blood. 2001;97(2):367-375
  163. 163. Miller JS, Rooney CM, Curtsinger J. et al. Expansion and homing of adoptively transferred human natural killer cells in immunodeficient mice varies with product preparation and in vivo cytokine administration: Implications for clinical therapy. Biology of Blood and Marrow Transplantation. 2014;20(8):1252-1257. DOI: 10.1016/j.bbmt.2014.05.004
  164. 164. Somanchi SS, Somanchi A, Cooper LJ, et al. Engineering lymph node homing of ex vivo‐expanded human natural killer cells via trogocytosis of the chemokine receptor CCR7. Blood. 2012;119(22):5164-5172. DOI: 10.1182/blood‐2011‐11‐389924
  165. 165. Kochenderfer JN, Wilson WH, Janik JE, et al. Eradication of B‐lineage cells and regression of lymphoma on patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116(20):4099-4102. DOI: 10.1182/blood‐2010‐04‐281931
  166. 166. Brentjens RJ, Rivière I, Park JH, et al. Safety and persistence of adoptively transferred autologous CD19‐targeted T cells in patients with relapsed or chemotherapy refractory B‐cell leukemias. Blood. 2001;118(18):4817-4828
  167. 167. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, Huls MH, Liu E, Gee AP, Mei Z, Yvon E, Weiss HL, Liu H, Rooney CM, Heslop HE, Brenner MK: Virus‐specificT cells engineered to coexpress tumor‐specific receptors:persistence and antitumor activityin individuals with neuroblastoma. Nature Medicine. 2008;14(11):1264-1270. DOI: 10.1038/nm.1882
  168. 168. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England Journal of Medicine. 2014;371(16):1507-1517. DOI: 10.1056/NEJMoa1407222
  169. 169. Morgan RA, Yang JC, Kitano M, et al. Case report of a serious adverse eventfollowing theadministration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Molecular Therapy. 2010;18:843-851. DOI: 10.1038/mt.2010.24
  170. 170. Müller T, Uherek C, Maki G, et al. Expression of a CD20‐specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK‐resistance of lymphoma and leukemia cells. Cancer Immunology, Immunotherapy. 2008;57(3):411-423. DOI: 10.1007/s00262‐007‐0383‐3
  171. 171. 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;106(1):376-383. DOI: 10.1182/blood‐2004‐12‐4797
  172. 172. Menard S, Pupa SM, Campiglio M, et al. Biologic and therapeutic role of HER2 in cancer. Oncogene. 2003;22(42):6570-6578. DOI: 10.1038/sj.onc.1206779
  173. 173. Meric‐Bernstam F, Hung MC. Advances in targeting human epidermal growth factor receptor‐2 signaling for cancer therapy. Clinical Cancer Research. 2006;12(21):6326-6330. DOI: 10.1158/1078‐0432.CCR‐06‐1732
  174. 174. Kruschinski A, Moosmann A, Poschke I, et al. Engineering antigen‐specific primary human NK cells against HER‐2 positive carcinomas. Proceedings of the National Academy of Sciences USA. 2008;105(45):17481-174816. DOI: 10.1073/pnas.0804788105
  175. 175. Uherek C, Tonn T, Uherek B, et al. Retargeting of natural killer‐cell cytolytic activity to ErbB2‐expressing cancer cells results in efficient and selective tumor cell destruction. Blood. 2002;100(4):1265-1273
  176. 176. Liu H, Yang B, Sun T, et al. Specific growth inhibition of ErbB2‐expressing human breast cancer cells by genetically modified NK‐92 cells. Oncology Reports. 2015;33(1):95-102. DOI: 10.3892/or.2014.3548
  177. 177. Chang YH, Connolly J, Shimasaki N, et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Research. 2013;73(6):1777-1786. DOI: 10.1158/0008‐5472.CAN‐12‐3558
  178. 178. Tassev DV, Cheng M, Cheung NK. Retargeting NK92 cells using an HLA‐A2‐restricted, EBNA3C‐specific chimeric antigen receptor. Cancer Gene Therapy. 2012;19(2):84-100. DOI: 10.1038/cgt.2011.66
  179. 179. Esser R, Müller T, Stefes D, et al. NK cells engineered to express a GD2‐specific antigen receptor display built‐in ADCC‐like activity against tumour cells of neuroectodermal origin. Journal of Cellular and Molecular Medicine. 2012;16(3):569-581. DOI: 10.1111/j.1582‐4934.2011.01343.x
  180. 180. Boissel L, Betancur M, Lu W, et al. Comparison of mRNA and lentiviral based transfection of natural killer cells with chimeric antigen receptors recognizing lymphoid antigens. Leukemia and Lymphoma. 2012;53(5):958-965. DOI: 10.3109/10428194.2011.634048
  181. 181. Chu J, Deng Y, Benson DM, et al. CS1‐specific chimeric antigen receptor (CAR)‐engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia. 2014;28(4):917-927. DOI: 10.1038/leu.2013.279
  182. 182. Jiang H, Zhang W, Shang P, Zhang H, Fu W, Ye F, et al. Transfection of chimeric anti‐CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Molecular Oncology. 2014;8(2):297-310. DOI: 10.1016/j.molonc.2013.12.001
  183. 183. Schönfeld K, Sahm C, Zhang C et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2‐specific chimeric antigen receptor. Molecular Therapy. 2015;23(2):330-338. DOI: 10.1038/mt.2014.219
  184. 184. Töpfer K, Cartellieri M, Michen S, et al. DAP12‐based activating chimeric antigen receptor for NK cell tumor immunotherapy. The Journal of Immunology2015;194(7):3201-3212. DOI: 10.4049/jimmunol.1400330
  185. 185. Altvater B, Landmeier S, Pscherer S, et al. 2B4 (CD244) signaling by recombinant antigen‐specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells. Clinical Cancer Research. 2009;15(15):4857-4866. DOI: 10.1158/1078‐0432.CCR‐08‐2810
  186. 186. Chu Y, Hochberg J, Yahr A, et al. Targeting CD20+ aggressive B‐cell Non‐Hodgkin lymphoma by anti‐CD20 CAR mRNA‐modified expanded natural killer cells in vitro and in NSG mice. Cancer Immunology Research. 2015;3(4):333-344. DOI: 10.1158/2326‐6066.CIR‐14‐0114

Written By

Yui Harada, Koji Teraishi, Minori Ishii, Hiroshi Ban and Yoshikazu Yonemitsu

Submitted: 02 December 2016 Reviewed: 05 April 2017 Published: 13 December 2017