Since its discovery in 1995, TNF-related apoptosis-inducing ligand (TRAIL) has sparked growing interest among oncologists due to its remarkable ability to induce apoptosis in malignant human cells, but not in most normal cells. However, one major drawback is its fast clearance rate in vivo. Thus, the development of an alternative means of delivery may increase the effectiveness of TRAIL-based therapy. In this study, we developed a secretory TRAIL-armed natural killer (NK) cell–based therapy and assessed its cytotoxic effects on colorectal cancer cells and its tumoricidal efficacy on colorectal peritoneal carcinomatosis xenograft. We generated genetically modified NK cells by transduction with a lentiviral vector consisting of a secretion signal domain, a trimerization domain, and an extracellular domain of the TRAIL gene. These NK cells secreted a glycosylated form of TRAIL fusion protein that induced apoptotic death. Intraperitoneally, but not intravenously, injected NK cells effectively accumulated at tumor sites, infiltrated tumor tissue, induced apoptosis, and delayed tumor growth. These results shed light on the therapeutic potential of genetically engineered NK cells to treat peritoneal carcinomatosis. Mol Cancer Ther; 15(7); 1591–601. ©2016 AACR.

Since preclinical studies in mice and primates have shown that administration of TNF-related apoptosis-inducing ligand (TRAIL) can induce apoptosis in human tumors without cytotoxicity to normal organs or tissue (1), Genentech and Amgen jointly prepared TRAIL and performed clinical trials on low-grade non-Hodgkin's lymphoma (2). Although the preclinical/clinical studies demonstrated the safety and activity of TRAIL, its short elimination half-life (approximately 30 minutes) is a main obstacle (3, 4). To overcome this obstacle, researchers have devised several strategies, including using humanized anti-TRAIL receptor antibodies (Ab; ref. 5–8) and a chimeric human TRAIL/human IgG-Fc fusion protein (9). In this study, we attempted to develop secretory TRAIL-natural killer (NK) cell–based therapy. Because NK cells possess tumor-infiltrating capabilities and accumulate selectively at tumor sites (10–13), we used NK cells as carriers to deliver secretory TRAIL.

NK cells are cytotoxic lymphocytes of the innate immune system that kill a variety of tumors and infected cells in the absence of previous stimulation in vivo and in vitro (14–16). NK cells possess two types of surface receptors: activating receptors (NCR, NKG2D, CD16, LY49) and inhibitory receptors [KIR (killer-cell immunoglobulin-like receptors), ILT/LIR, LY49, PD-1; refs. 17–22]. NK-cell activation results from the balance of signals produced by these two receptors. NK cells are activated by several cytokines, such as IL2, IL12, IL15, IL18, IL21, and IFNα/β (23, 24). Upon activation, NK cells proliferate and upregulate effector molecules, such as perforin, granzymes, Fas ligand (FasL), TNFα, and TRAIL (24–27). In contrast, Velthuis and colleagues reported that IL2-activated NK cells induce apoptosis by secretion of granzyme B and perforin, but not via the FasL, TNFα, or TRAIL pathways (28). NK cells can play an important role in immunosurveillance of tumors by directly inducing the apoptotic death of tumor cells (29). These observations support that the mechanism of NK cytotoxicity mainly relies on secretory granules, granzyme B, and requires cell adhesion (22, 30). NK cells also have an immunoregulatory role as they secrete several cytokines, such as IFNγ, following their ligand interaction with cell-surface receptors (31).

Moreover, NK cells demonstrate the ability to infiltrate tumors (10, 11). Because NK cells can recognize tumor cells and infiltrate solid tumors, one of the main goals of this study was to develop secretory TRAIL-armed IL2-activated NK (A-NK) cells and assess their tumoricidal efficacy in in vitro and in vivo systems.

In this study, we constructed pLenti-FETZ vector, which contains three functional domains: a secretion signal domain (the extracellular domain of a ligand for Flt3 tyrosine kinase receptor), a leucine zipper domain for trimerization, and the extracellular domain of TRAIL (a.a. 95-281). NK-92MI-FETZ cells were generated via lentiviral transduction; they can secrete high levels of glycosylated TRAIL fusion protein and induce cell death and apoptosis in colorectal cancer cell lines. Notably, NK-92MI-FETZ cells can infiltrate mouse peritoneal tumors and inhibit peritoneal tumor growth in vivo, which sheds light on their therapeutic potential for the treatment of colorectal peritoneal carcinomatosis.

Cell cultures

Human colorectal carcinoma CX-1 cells were obtained from Dr. J.M. Jessup (National Institutes of Health, Bethesda, MD) in 1999, and no authentification was performed by the authors. They were cultured in RPMI-1640 medium (Gibco BRL) containing 10% FBS (HyClone). Human colorectal cancer HCT116 and LS174T cells were purchased from the American Tissue Type Culture Collection (ATCC) in 2014, and no authentification was performed by the authors. They were cultured in McCoy's 5A medium or DMEM (Gibco-BRL) containing 10% FBS. NK-92MI cells were purchased from the ATCC in 2014, and no authentification was done by the authors. They were cultured in minimum essential medium-α (Life Technologies) supplemented with 2 mmol/L l-glutamine, 0.2 mmol/L inositol, 0.1 mmol/L 2-mercaptoethanol, 0.02 mmol/L folic acid, 12.5% horse serum, and 12.5% FBS. HEK293T cells were purchased from the ATCC in 2014, and no authentification was done by the authors. They were cultured in DMEM supplemented with 10% heat-inactivated FBS.

Reagents and antibodies

Anti-caspase 8, anti-caspase 9, anti-caspase 3, anti-PARP, and anti-human CD45 Ab were purchased from Cell Signaling Technology. Anti-TRAIL Ab was obtained from Santa Cruz Biotechnology. For production of TRAIL, a human TRAIL cDNA fragment (amino acids 114–281) obtained via RT-PCR was cloned into a pET-23d (Novagen) plasmid, and His-tagged TRAIL protein was purified using the Qiagen express protein purification system (Qiagen).

MTS assays

[3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) studies were carried out using the Promega CellTiter 96 AQueous One Solution Cell Proliferation Assay (Promega). Cells (1 × 105) were grown in RPMI-1640 medium containing 10% FBS in tissue culture–coated 96-well plates and treated with drugs for 24 hours. Cells were then treated with 20 μL MTS/phenazine methosulfate solution for 1 to 4 h at 37°C. Absorbance at 490 nm was determined using an ELISA plate reader.

Construction of Lenti-FETZ vector and transfection into NK-92 cells

pFETZ was obtained from Dr. Y He (Immunotherapy Center, Medical College of Georgia, GA). It contains the extracellular domain of Flt3L (a.a. 1–81), an isoleucine zipper domain, and the extracellular domain of TRAIL (a.a. 95–281). pFETZ was subcloned into pENTER-D-TOPO (Invitrogen) as an entry clone. In vitro recombination between an entry clone (containing a gene of interest flanked by attL sites) and a destination vector was performed to construct pLenti-FETZ/GFP expression vector. Clones with the right sequence were chosen. Lentivirus carrying a secretable trimeric TRAIL gene is called Lenti-FETZ, and Lenti-GFP virus served as a control.

Lentiviral particles are generated by transfection of the following plasmids [the control plasmid pLenti-GFP or the expression plasmid (i.e., pLenti-FETZ), plus pLenti-3A, pLenti-3B, and pLenti-3C] into 293-T cells using Lipofectamine 2000 (Life technologies). Culture media were harvested 48 hours after transfection, filtered through 0.45 μm filters, underwent ultracentrifugation at 100,000 × g for 2 hours at 4°C, and were stored at −80°C in single-use aliquots.

NK-92MI cells were transduced with the lentivirus (Lenti-GFP and Lenti-FETZ). Multiplicity of infection (MOI) was between 20 and 100. Upon infection, NK-92MI cells were selected with 2 μg/mL puromycin for 3 weeks.

Analysis of glycosylated secretory TRAIL protein

Glycosylation of secreted TRAIL was examined by treatment with three different types of glycosidases. It is well known that O-Glycosidase can remove desialylated core 1 and core 3 O-linked disaccharides attached to Ser/Thr residues. Endo H is a recombinant glycosidase and can remove only high-mannose and some hybrid types of N-linked carbohydrates. Unlike Endo H, PNGase F can remove all types of N-linked (Asn linked) glycosylation regardless of their types (high-mannose, hybrid, bi-, tri-, and tetra-antennary). Supernatant of NK-92MI-FETZ was treated with three different types of glycosidases, and then glycosylated and deglycosylated TRAIL were determined by immunoblotting assay.

Immunoblot analysis

Protein was measured with BCA Protein Assay Reagent (Pierce) and separated with SDS-PAGE gel and transferred to nitrocellulose membrane. The membrane was then blocked with 5% nonfat dry milk in TBS-Tween-20 for 0.5 hours and incubated with primary Ab at 4°C overnight. The membrane was incubated with horseradish peroxidase–conjugated anti-rabbit or anti-mouse IgG at room temperature for 1 hour and then visualized using the chemiluminescence protocol.

ELISA

The supernatant of each NK cell culture was collected and examined using ELISA to measure the concentrations of soluble TRAIL. The supernatants of the NK cell culture and cell protein extract were centrifuged for 10 minutes at 6,000 x g and analyzed with an ELISA kit (R&D systems) to determine the concentrations of TRAIL.

Flow cytometry

Single-cell suspensions were stained with FITC- or allophycocyanin (APC)-conjugated CD45 Abs. To distinguish NK-92 cells from tumor cells, cell surface marker human CD45 was used. The conjugated Ab-specific to human CD45 was obtained from BioLegend. HCT116 cells have no expression of CD45, whereas NK-92MI cells are strongly positive (Supplementary Fig. S1B). An Annexin-V–FITC Apoptosis Detection kit (BD Pharmingen) was used to measure apoptosis. HCT116, NK-92MI, and NK-92MI-FETZ cells were stained with propidium iodide (PI) and FITC-conjugated Annexin V and analyzed with flow cytometry (Supplementary Fig. S1C). Data were collected on a FACSCalibur (BD Biosciences) and analyzed using CellQuest Pro software (BD Biosciences).

51Cr release assay

Cytolytic activity was assessed using a standard 51Cr release assay. Target cells (HCT116) were labeled with 50 μCi 51Cr, and 1 × 103 cells/well were incubated in a U-bottom 96-well plate with effector cells (NK-92MI), at indicated conditions, at the indicated effector/target (E/T) ratios. After 4 hours, 50 μL aliquots of cell-free supernatant were harvested and assayed for 51Cr release in TopCounter reader (PerkinElmer) and then analyzed using the following formula: (test 51Cr release – spontaneous 51Cr release)/(maximal 51Cr release − spontaneous 51Cr release) × 100. Spontaneous 51Cr release represents the amount of 51Cr released by target cells in the absence of effector cells. The maximal 51Cr release was determined by measuring the 51Cr radioactivity from complete lysis of target cells with 1% Triton X-100.

Cell migration assay

An in vitro cell migration assay was performed to determine the tropism of NK-92MI cells for cancer cell–conditioned medium. Cancer cell culture supernatant (600 μL) was added in the bottom well of a Transwell plate (5-μm pore membrane; Corning) before 100,000 NK-92MI-GFP or NK-92MI-FETZ cells in 100 μL were added to the top well. NK-92MI cells were allowed to migrate across the membrane for 4 hours at 37°C. The cells in the lower chamber were counted.

In vivo imaging of NK-92MI homing

LS174T cells (1 × 106) expressing firefly luciferase were delivered i.p. NK-92MI cells (1 × 106) were labeled using Cy5.5 NHS ester (Lumiprobe Corporation) for 30 minutes before injection, according to the manufacturer's instructions, and delivered either by i.v. or i.p. injections 7 days after tumor cell injection. Bioluminescence and fluorescence were determined using an in vivo imaging system (IVIS200 system, Perkin Elmer). At study termination, mice were sacrificed and samples of tumors, serums, peritoneal washes, spleens, and livers were collected for further analysis.

Immunohistochemistry

Fixed specimens were embedded in paraffin and cut into 4-μm sections for hematoxylin and eosin staining. GFP Ab (rabbit polyclonal; Invitrogen) and TRAIL Ab (rabbit monoclonal; Santa Cruz Biotechnology) were used as primary Abs and detected with biotinylated secondary Abs and diaminobenzidine (Vector Laboratories).

Fluorescent microscopy was used to detect Cy5.5-positive cells with 4′,6-diamidino-2-phenylindole (DAPI) counterstain. Terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling (TUNEL) staining was performed according to the manufacturer's instructions (Roche Diagnostics). Microscopy was conducted using a light (Olympus BX40), fluorescent (Carl Zeiss; Axioskop 2), or confocal (Bio-Rad MRC 1024) microscope.

Animal model

NOD.CB17-Prkdcscid/J mice (six weeks old, Taconic) were challenged i.p. with 1 × 106 LS174T cells expressing firefly luciferase, which was stably transduced by lentiviral transfection of the pGL4 Luciferase Reporter Vector (Promega) and selected with puromycin. The luciferase signal was monitored by injecting the luciferase substrate luciferin (150 mg/kg, i.p.; GoldBio) 5 minutes after anesthesia with 2% isoflurane prior to imaging on an IVIS200 system. Bioluminescence signal was quantified using the LivingImage software (Perkin Elmer). The mice were examined for bioluminescent signal, indicating basal tumor loading. Mice with the same level of bioluminescence were divided into four groups (seven per group) and given treatments of PBS, NK-92MI-GFP (5 × 106, i.p.), or NK-92MI-FETZ (5 × 106, i.p.) twice per week for 2 weeks. The tumor load, represented by bioluminescent signal determined using the IVIS bioluminescent Imaging System, was checked twice a week. Mice were fed ad libitum and maintained in environments with a controlled temperature of 22 to 24°C and 12-hour light and dark cycles. All animal experiments were carried out at the University of Pittsburgh in accordance with the Guide for the Care and Use of Laboratory Animals.

Statistical analysis

Statistical analysis was performed using GraphPad Prism 5 (GraphPad Software). In vivo experiments with multiple groups were analyzed using repeated measures ANOVA, and single group data were assessed using the Student t test. Differences were considered statistically significant when the P value was <0.05. All in vitro experiments were performed in triplicate unless specified.

NK-92MI cells are resistant to TRAIL-mediated cytotoxicity

To investigate whether TRAIL is cytotoxic to NK-92MI cells, we measured cell viability with MTS assay. NK-92MI is an IL2-independent NK-cell line derived from a male patient with rapidly progressive non-Hodgkin lymphoma (32). HCT116/LS174T/NK92MI cells were treated with various concentrations of TRAIL, and then viability was determined. As shown in Fig. 1A, HCT116 and LS174T cell viability decreased as TRAIL concentration increased. HCT116 cells were more sensitive than LS174T cells to TRAIL. However, unlike HCT116 and LS174T cells, NK-92MI cells were resistant to TRAIL. Similar results were obtained in terms of TRAIL-induced PARP cleavage, the hallmark of apoptosis (Fig. 1B). These results demonstrated that NK-29MI cells can be used as carriers to deliver secretory TRAIL.

Figure 1.

NK-92MI cells are resistant to TRAIL-mediated cytotoxicity. A, human colorectal carcinoma HCT116, human colorectal adenocarcinoma LS174T, and human IL2-independent NK-92MI cells were seeded at 104 cells per well and grown overnight. Cells were treated with various concentrations (0–1,500 ng/mL) of human recombinant TRAIL (rTRAIL) for 24 hours, and MTS assay was performed to measure viability. Error bars, SD from triplicate experiments. *, P < 0.05; **, P < 0.01, statistically significant difference compared with the control. B, HCT116, LS174T, and NK-92MI cells were treated with rTRAIL (0–500 ng/mL) for 24 hours, and PARP was detected by immunoblotting. Actin was used to confirm that equal amounts of proteins were loaded in each lane.

Figure 1.

NK-92MI cells are resistant to TRAIL-mediated cytotoxicity. A, human colorectal carcinoma HCT116, human colorectal adenocarcinoma LS174T, and human IL2-independent NK-92MI cells were seeded at 104 cells per well and grown overnight. Cells were treated with various concentrations (0–1,500 ng/mL) of human recombinant TRAIL (rTRAIL) for 24 hours, and MTS assay was performed to measure viability. Error bars, SD from triplicate experiments. *, P < 0.05; **, P < 0.01, statistically significant difference compared with the control. B, HCT116, LS174T, and NK-92MI cells were treated with rTRAIL (0–500 ng/mL) for 24 hours, and PARP was detected by immunoblotting. Actin was used to confirm that equal amounts of proteins were loaded in each lane.

Close modal

Development and characterization of secretory TRAIL-armed NK-92MI cells

We constructed a lentiviral vector (pLenti-FETZ) producing a secretable form of TRAIL or pLenti-GFP as a control lentiviral vector (Fig. 2A). We chose lentiviral vectors because they are able to efficiently transduce NK-92MI cells (Fig. 2B). We established NK-92MI-GFP and NK-92MI-FETZ stable cell lines by selecting the lentiviral vector–infected NK-92MI cells with puromycin treatment for 3 weeks. Interestingly, we observed an expression of 40 kD TRAIL fusion protein in NK-92MI-FETZ cell lysate as well as 50 kD TRAIL fusion protein in the culture medium (Fig. 2C). These results suggest that the chimeric gene FETZ was expressed, modified, and secreted into the medium. To examine whether secreted TRAIL fusion protein was modified, three different types of glycosidases were treated to inhibit glycosylation of secreted TRAIL in the supernatant of NK92MI-FETZ cell–cultured medium. The molecular weight decreased after PNGase F (N-Glycosidase F) treatment, which was able to remove the N-linked glycosylation, but not O-Glycosidase and Endoglycosidase H (Endo H; Fig. 2D). The results from Fig. 2D indicate that secretory TRAIL contains N-linked Endo H insensitive carbohydrates. To quantify the level of secreted TRAIL from NK-92MI-FETZ cells, ELISA was performed. We observed that the level of secreted TRAIL gradually increased as a function of time (Fig. 2E). Results from immunoblot assay and ELISA demonstrate that NK-92MI-FETZ cells are able to secrete substantial amounts of TRAIL fusion protein.

Figure 2.

Development and characterization of secretory TRAIL-armed NK-92MI cells. A, schematic lentivirus vector maps of the recombinant DNA constructs were drawn. To deliver secretory TRAIL to tumor, we created a lentiviral vector (Lenti-FETZ) producing a secretable form of TRAIL by fusing coding sequences for the secretory domain of Flt3L (a.a. 1–81) and an isoleucine zipper with the extracellular domain of TRAIL (a.a. 95–281). We also created a lentiviral vector (Lenti-GFP)–producing GFP. B, to determine infection efficiency, Lenti-GFP vector was transduced into NK-92MI at an MOI of 40. After 2 days, phase-contrast image or fluorescence image was visualized by light or fluorescence microscopy, respectively. C, to measure production of TRAIL, NK-92MI cells were stably transduced with Lenti-FETZ. TRAIL expression (cell lysate) and secreted TRAIL (conditioned medium) in NK-92MI-FETZ cells were determined with Western blot analysis. D, to examine translational modification of secreted TRAIL protein, glycosidases (O-Glycosidase, PNGase F, and Endo H) were added to inhibit glycosylation of secreted TRAIL in the supernatant of NK-92MI-FETZ cells. Glycosylated and deglycosylated TRAIL were determined using Western blot analysis. E, to measure amounts of secreted TRAIL, 2 × 104 NK-92MI-GFP and NK-92MI-FETZ cells were seeded in 96 wells, and the supernatants were harvested at the indicated time. The concentration of secreted TRAIL was quantified using ELISA assay. Error bars, SE from triplicate groups.

Figure 2.

Development and characterization of secretory TRAIL-armed NK-92MI cells. A, schematic lentivirus vector maps of the recombinant DNA constructs were drawn. To deliver secretory TRAIL to tumor, we created a lentiviral vector (Lenti-FETZ) producing a secretable form of TRAIL by fusing coding sequences for the secretory domain of Flt3L (a.a. 1–81) and an isoleucine zipper with the extracellular domain of TRAIL (a.a. 95–281). We also created a lentiviral vector (Lenti-GFP)–producing GFP. B, to determine infection efficiency, Lenti-GFP vector was transduced into NK-92MI at an MOI of 40. After 2 days, phase-contrast image or fluorescence image was visualized by light or fluorescence microscopy, respectively. C, to measure production of TRAIL, NK-92MI cells were stably transduced with Lenti-FETZ. TRAIL expression (cell lysate) and secreted TRAIL (conditioned medium) in NK-92MI-FETZ cells were determined with Western blot analysis. D, to examine translational modification of secreted TRAIL protein, glycosidases (O-Glycosidase, PNGase F, and Endo H) were added to inhibit glycosylation of secreted TRAIL in the supernatant of NK-92MI-FETZ cells. Glycosylated and deglycosylated TRAIL were determined using Western blot analysis. E, to measure amounts of secreted TRAIL, 2 × 104 NK-92MI-GFP and NK-92MI-FETZ cells were seeded in 96 wells, and the supernatants were harvested at the indicated time. The concentration of secreted TRAIL was quantified using ELISA assay. Error bars, SE from triplicate groups.

Close modal

Apoptotic activity of secretory TRAIL-armed NK cells in noncontacting (Transwell) coculture system

Next, we investigated whether NK-92MI-FETZ cells are able to kill cancer cells. We used the Transwell assay as a noncontacting coculture system to evaluate their cytotoxic effect. As shown in Fig. 3A, HCT116 cells demonstrated clear morphologic features typical of apoptotic death, such as shrinkage and blebbing, when NK-92MI-FETZ cells, but not NK-92MI cells, were cocultured for 24 hours. We also confirmed that secretory TRAIL-armed NK-92MI-FETZ cells and conditioned medium from these cells remarkably induced PARP cleavage (the hallmark feature of apoptosis) and activation of caspases in HCT116 cells (lanes 4 and 6 in Fig. 3B). Unlike secretory TRAIL-armed NK-92MI-FETZ cells, unarmed NK-92MI cells and conditioned medium from these cells induced minimal apoptosis in HCT116 cells (lanes 3 and 5 in Fig. 3B). As shown in Fig. 3B and C, there is not much difference between NK-92MI cells and NK-92MI-GFP cells. These data clearly demonstrate that secretory TRAIL from NK-92MI-FETZ cells effectively induces cytotoxicity in HCT116 cells. Similar results were observed in LS174T cells, but we incubated NK-92MI-FETZ cells with LS174T cells for 48 hours to see the apoptotic cell death, indicating that LS174T cells are less sensitive to secretory TRAIL than HCT116 cells (Fig. 3D). Data from Annexin V/PI assay confirmed secretory TRAIL-induced apoptosis in HCT116 cells (Fig. 3E; apoptotic death cells in top right plot quadrants). Apoptosis occurred in a NK-92MI-FETZ cell number (secretory TRAIL dose)–dependent manner. Results from Western blot assay also confirmed that apoptosis occurred in a secretory TRAIL dose–dependent manner (Fig. 3F).

Figure 3.

Apoptotic activity of secretory TRAIL-armed NK cells in a noncontacting (Transwell) coculture system. A and B, HCT116 cells were seeded in the lower chamber of the Transwell (pore size: 0.4 μm); NK-92MI or NK-92MI-FETZ cells were plated in the upper chamber. The Transwell chamber was incubated for 24 hours. Morphology of HCT116 cells in the lower chamber was examined under a light microscope (A). Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of HCT116 cells in the lower chamber, which were cocultured with NK-92MI or NK-92MI-FETZ cells in the upper chamber or their conditioned medium for 24 hours (B). C, HCT116 cells in the lower chamber were incubated with NK-92MI, NK-92MI-GFP, or NK-92MI-FETZ cells in the upper chamber or their conditioned medium for 24 hours. Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of HCT116 cells were examined. D, LS174T cells plated in the lower chamber were cocultured with NK-92MI-GFP or NK-92MI-FETZ cells plated in the upper chamber or their conditioned medium for 48 hours. Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of LS174T cells were examined. Actin was used as an internal standard. E, HCT116 cells plated in the lower chamber were cocultured with NK-92MI-GFP or NK-92MI-FETZ cells plated in the upper chamber for 24 hours. HCT116 cells were stained with FITC–Annexin V and PI. Apoptosis was detected with flow cytometric assay (arrows). F, HCT116 cells plated in the lower chamber were cocultured with various numbers of NK-92MI-GFP or NK-92MI-FETZ cells plated in the upper chamber for 24 hours. Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of HCT116 cells were examined. Actin was shown as an internal standard.

Figure 3.

Apoptotic activity of secretory TRAIL-armed NK cells in a noncontacting (Transwell) coculture system. A and B, HCT116 cells were seeded in the lower chamber of the Transwell (pore size: 0.4 μm); NK-92MI or NK-92MI-FETZ cells were plated in the upper chamber. The Transwell chamber was incubated for 24 hours. Morphology of HCT116 cells in the lower chamber was examined under a light microscope (A). Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of HCT116 cells in the lower chamber, which were cocultured with NK-92MI or NK-92MI-FETZ cells in the upper chamber or their conditioned medium for 24 hours (B). C, HCT116 cells in the lower chamber were incubated with NK-92MI, NK-92MI-GFP, or NK-92MI-FETZ cells in the upper chamber or their conditioned medium for 24 hours. Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of HCT116 cells were examined. D, LS174T cells plated in the lower chamber were cocultured with NK-92MI-GFP or NK-92MI-FETZ cells plated in the upper chamber or their conditioned medium for 48 hours. Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of LS174T cells were examined. Actin was used as an internal standard. E, HCT116 cells plated in the lower chamber were cocultured with NK-92MI-GFP or NK-92MI-FETZ cells plated in the upper chamber for 24 hours. HCT116 cells were stained with FITC–Annexin V and PI. Apoptosis was detected with flow cytometric assay (arrows). F, HCT116 cells plated in the lower chamber were cocultured with various numbers of NK-92MI-GFP or NK-92MI-FETZ cells plated in the upper chamber for 24 hours. Immunoblot analysis of caspase 8/9/3 activation and PARP cleavage of HCT116 cells were examined. Actin was shown as an internal standard.

Close modal

Apoptotic activity of secretory TRAIL-armed NK cells in contacting culture system

To examine the cytotoxic effect of NK-92MI-FETZ cells on colorectal cancer cells in a contacting culture system, CX-1 cells were cocultured with NK-92MI-FETZ cells in a cell culture dish. As shown in Fig. 4A, detachment of GFP stably transfected CX-1 cells was observed, and a high ratio of effector to target (E:T) cells increased the detachment of CX-1 cells (Fig. 4A). We observed that NK-92MI-FETZ cells were more effective than NK-92MI cells at inducing the detachment of CX-1 cells. Similar results were observed when HCT116 cells were cocultured with NK-92MI/NK-92MI-FETZ cells (Supplementary Fig. S1A). To quantitatively analyze this, HCT116 cells and NK-92MI/NK-92MI-FETZ were cocultured with various ratios, and cells that underwent apoptotic death were detected with flow cytometric analysis (Fig. 4B). Using CD45 staining, the tumor cells were able to be gated as non-CD45 cells when tumor cells were mixed with NK-92MI cells (left plots in Fig. 4B). After mixing NK-92MI or NK-92MI-FETZ with HCT116 cells at an increased E:T ratio, HCT116 cells underwent increased apoptotic cell death (early apoptotic death cells in lower right plot quadrants, Annexin V+ PI and late apoptotic death cells in upper right plot quadrants, Annexin V+ PI+) and necrosis (upper left plot quadrants: Annexin V PI+) in NK-92MI-FETZ–treated tumor cells compared with NK-92MI cells (middle and right plots in Fig. 4B). We also used luciferase quantitative assay to measure tumor-specific NK-cell cytotoxic activity. Reduction in tumor cell–associated luciferase activity closely reflected the killing activity of the NK cells against tumor targets. NK-92MI-GFP or NK-92MI-FETZ cells were incubated with luciferase cDNA stably transfected HCT116-luc+ cells at a ratio of 4:1 for 24 hours. The luciferase activity of NK-92MI-FETZ cells mixed with HCT116 cells was significantly decreased compared with that of NK-92MI-GFP cells (Fig. 4C). Similar results were observed with 51Cr release assay (Fig. 4D). Moreover, NK-92MI-FETZ–induced cytotoxicity was suppressed by treatment with neutralizing anti-TRAIL Ab. These results revealed that enhanced cytotoxic effects of NK-92MI-FETZ are due to secretion of TRAIL. In summary, results from microscopic examination, flow cytometric analysis, luciferase quantitative assay, and 51Cr release assay demonstrate that NK-92MI-FETZ cells have more cytotoxic efficacy than NK-92MI cells.

Figure 4.

Apoptotic activity of secretory TRAIL-armed NK cells in a contacting culture system. A, GFP stably transfected CX-1 (CX-1-GFP) cells (target, T) were seeded and grown overnight and then contact cocultured with NK-92MI/FETZ cells (effector, E) at various E:T ratios. After 24 hours, phase-contrast image or fluorescence image was visualized with light or fluorescence microscopy, respectively. B, HCT116 cells and NK-92MI or NK-92MI-FETZ cells were contact cocultured at various E:T ratios. After 24 hours, cells were stained with anti-human CD45-APC antibody, FITC–Annexin V, and PI. CD45 status (left plots) and apoptosis of CD45-negative HCT116 cells (middle and right plots) were analyzed using flow cytometry. C, HCT116-luc+ cells were plated in triplicate wells and contact cocultured without or with NK-92MI-GFP or NK-92MI-FETZ cells (E:T ratio at 4:1). After 24 hours, luciferase activity was measured. Error bars, SE from triplicates. **, P < 0.01, statistically significant difference. D, 51Cr-labeled HCT116 cells were plated in triplicate wells and contact cocultured without or with NK-92MI-GFP or NK-92MI-FETZ cells (E:T ratio at 4:1) in the presence or absence of 2 μg/mL anti-TRAIL antibody or mouse IgG1. Cytotoxicity is expressed as the mean percentage specific lysis and SDs. Statistically significant differences between HCT116 and HCT116+NK-92MI-GFP (*, P < 0.05), HCT116+NK-92MI-GFP and HCT116+NK-92MI-FETZ (++, P < 0.01), and HCT116+NK-92MI-FETZ and HCT116+NK-92MI-FETZ+anti-TRAIL antibody ($$, P < 0.01) are shown.

Figure 4.

Apoptotic activity of secretory TRAIL-armed NK cells in a contacting culture system. A, GFP stably transfected CX-1 (CX-1-GFP) cells (target, T) were seeded and grown overnight and then contact cocultured with NK-92MI/FETZ cells (effector, E) at various E:T ratios. After 24 hours, phase-contrast image or fluorescence image was visualized with light or fluorescence microscopy, respectively. B, HCT116 cells and NK-92MI or NK-92MI-FETZ cells were contact cocultured at various E:T ratios. After 24 hours, cells were stained with anti-human CD45-APC antibody, FITC–Annexin V, and PI. CD45 status (left plots) and apoptosis of CD45-negative HCT116 cells (middle and right plots) were analyzed using flow cytometry. C, HCT116-luc+ cells were plated in triplicate wells and contact cocultured without or with NK-92MI-GFP or NK-92MI-FETZ cells (E:T ratio at 4:1). After 24 hours, luciferase activity was measured. Error bars, SE from triplicates. **, P < 0.01, statistically significant difference. D, 51Cr-labeled HCT116 cells were plated in triplicate wells and contact cocultured without or with NK-92MI-GFP or NK-92MI-FETZ cells (E:T ratio at 4:1) in the presence or absence of 2 μg/mL anti-TRAIL antibody or mouse IgG1. Cytotoxicity is expressed as the mean percentage specific lysis and SDs. Statistically significant differences between HCT116 and HCT116+NK-92MI-GFP (*, P < 0.05), HCT116+NK-92MI-GFP and HCT116+NK-92MI-FETZ (++, P < 0.01), and HCT116+NK-92MI-FETZ and HCT116+NK-92MI-FETZ+anti-TRAIL antibody ($$, P < 0.01) are shown.

Close modal

Selective accumulation of NK-92MI cells at tumor sites in vitro and in vivo

Several researchers have reported that NK cells possess tumor-infiltrating capability and accumulate selectively at tumor sites (10–13). First, we investigated whether NK-92MI or NK-92MI-FETZ cells can migrate to cancer cells in vitro. We harvested conditioned medium from CX-1/LS174T/HCT116 cells and added them into the lower chamber of the Transwell plate and seeded NK-92MI-GFP/NK-92MI-FETZ cells into the upper chamber. We observed that migration of NK-92MI cells significantly increased to the conditioned medium from tumor cells in comparison to medium alone (Fig. 5A). No differences were observed regarding the migration ability of NK-92MI-GFP and NK-92MI-FETZ cells (Fig. 5A). Second, we investigated whether NK-92MI cells can infiltrate peritoneal carcinomatosis in vivo. To establish xenograft tumor formation, we i.p. injected LS174T-luc+ cells into NOD.CB17-Prkdcscid/J mice. These mice are characterized by an absence of functional T cells and B cells and have low NK-cell activity. We compared the efficiency of i.p. versus i.v. delivery of NK-92MI cells to the xenograft tumor. Figure 5B shows an accumulation of NK-92MI cells at tumor sites; i.p. injection was more effective than i.v. injection for delivery of NK-92MI cells. To confirm these observations, tumor tissues were removed, sectioned, and stained with DAPI. DAPI (tumor cell) and Cy5.5 (NK-92MI cell) fluorescent-stained cells were detected. Tumor-infiltrating NK-92MI cells were detected in i.p.-injected mice, but not many were detected in i.v.-injected mice (Fig. 5C). Time sequence examinations confirmed these results. Figure 5D shows that the Cy5.5 fluorescence signal radiance in the i.v. group was less than that of the i.p. group at every time point. To examine accumulation of NK-92MI cells in various tissues, tumor-bearing mice were i.p. or i.v. injected with NK-92MI-Cy5.5 cells, and the presence of NK-92MI-Cy5.5 cells was examined in the liver, spleen, lymph nodes, and tumor tissues. We observed that NK-92MI cells had mainly accumulated in the liver with i.v. injection, but not much in tumor tissues, whereas NK-92MI cells had accumulated in the liver, lymph nodes, and tumor tissues with i.p. injection (Fig. 5E). We also collected blood and peritoneal wash from i.p.- or i.v.-injected mice. The CD45 marker was used to distinguish human NK-92MI cells from the mouse-originated cells. We observed that human CD45-expressing cells (NK-92MI) were present in both the peritoneal wash and blood in both groups. There was 15-fold more NK-92MI cells in the peritoneal wash in the i.p. group than in the i.v. group (Fig. 5F and G). All these data confirmed that NK-92MI cells were able to infiltrate the LS174T peritoneal tumor, and that i.p. injection was more effective than i.v. injection at delivering NK-92MI cells to tumors.

Figure 5.

Selective accumulation of NK-92MI cells at tumor sites in vitro and in vivo. A, conditioned medium of CX-1/LS174T/HCT116 cells was added in the lower chamber of the Transwell plate (pore size: 5 μm). 1 × 105 NK-92MI-GFP or NK-92MI-FETZ cells were seeded in the upper chamber. After 4 hours, cells in the lower chamber were counted. Error bars, SE from triplicate experiments. *, P < 0.05; **, P < 0.01, statistically significant differences compared with the medium control group. B–F, NOD.CB17-Prkdcscid/J mice were i.p. inoculated with 5 × 105 LS174T cells expressing luciferase (LS174T-luc+). At day 7 of tumor growth, 5 × 106 Cy5.5 dye-stained NK-92MI (NK-92MI-Cy5.5) cells were injected i.p. or i.v. B, after 24 hours, mice were anesthetized (2% isoflurane) prior to imaging for Cy5.5 fluorescence at 670 to 680 nm and then injected i.p. with luciferin (30 mg/kg) for imaging bioluminescent activity to measure tumor loading. Luciferase signal (top plots) and Cy5.5 signal (bottom plots) were monitored using the IVIS Imaging System Series 200. C, after 48 hours of NK-92MI-Cy5.5 cell injection, tumor tissues were harvested, sectioned, and stained with DAPI. DAPI (tumor cell) and Cy5.5 (NK-92MI cell) fluorescent-stained cells were detected. D, the presence of i.p.-injected NK-92MI cells vs. i.v.-injected NK-92MI cells was examined various times after injection. Cy5.5 signals were plotted at different time points. E, NK-92MI-Cy5.5 cells were i.p. or i.v. injected. After 48 hours, tumor (LS174T-luc+)-bearing mice were sacrificed, and the liver, spleen, some lymph nodes, and tumor tissue were harvested (three mice/group). The presence of NK-92MI cells (Cy5.5 signals) and tumor cells (luciferase signals) at each organ was detected with the IVIS system. Effect of i.p. injection and i.v. injection on accumulation of NK-92MI at the tissues was also compared. Representative photos are shown. F and G, after 48 hours of NK-92MI injection, the presence of i.p.-injected vs. i.v.-injected NK-92MI cells was examined by detecting CD45-positive NK-92MI cells in the blood and peritoneal wash with flow cytometry (F) and plotting data from three mice each group (G). *, P < 0.05, statistically significant difference compared with the sham group.

Figure 5.

Selective accumulation of NK-92MI cells at tumor sites in vitro and in vivo. A, conditioned medium of CX-1/LS174T/HCT116 cells was added in the lower chamber of the Transwell plate (pore size: 5 μm). 1 × 105 NK-92MI-GFP or NK-92MI-FETZ cells were seeded in the upper chamber. After 4 hours, cells in the lower chamber were counted. Error bars, SE from triplicate experiments. *, P < 0.05; **, P < 0.01, statistically significant differences compared with the medium control group. B–F, NOD.CB17-Prkdcscid/J mice were i.p. inoculated with 5 × 105 LS174T cells expressing luciferase (LS174T-luc+). At day 7 of tumor growth, 5 × 106 Cy5.5 dye-stained NK-92MI (NK-92MI-Cy5.5) cells were injected i.p. or i.v. B, after 24 hours, mice were anesthetized (2% isoflurane) prior to imaging for Cy5.5 fluorescence at 670 to 680 nm and then injected i.p. with luciferin (30 mg/kg) for imaging bioluminescent activity to measure tumor loading. Luciferase signal (top plots) and Cy5.5 signal (bottom plots) were monitored using the IVIS Imaging System Series 200. C, after 48 hours of NK-92MI-Cy5.5 cell injection, tumor tissues were harvested, sectioned, and stained with DAPI. DAPI (tumor cell) and Cy5.5 (NK-92MI cell) fluorescent-stained cells were detected. D, the presence of i.p.-injected NK-92MI cells vs. i.v.-injected NK-92MI cells was examined various times after injection. Cy5.5 signals were plotted at different time points. E, NK-92MI-Cy5.5 cells were i.p. or i.v. injected. After 48 hours, tumor (LS174T-luc+)-bearing mice were sacrificed, and the liver, spleen, some lymph nodes, and tumor tissue were harvested (three mice/group). The presence of NK-92MI cells (Cy5.5 signals) and tumor cells (luciferase signals) at each organ was detected with the IVIS system. Effect of i.p. injection and i.v. injection on accumulation of NK-92MI at the tissues was also compared. Representative photos are shown. F and G, after 48 hours of NK-92MI injection, the presence of i.p.-injected vs. i.v.-injected NK-92MI cells was examined by detecting CD45-positive NK-92MI cells in the blood and peritoneal wash with flow cytometry (F) and plotting data from three mice each group (G). *, P < 0.05, statistically significant difference compared with the sham group.

Close modal

Effect of NK-92MI-FETZ on the growth of LS174T intraperitoneal tumors

To investigate the tumoricidal efficacy of TRAIL, we investigated the effect of NK-92MI-FETZ on the growth of LS174T intraperitoneal tumors in vivo. Figure 6B and C show that administration with NK-92MI-FETZ cells significantly delayed tumor growth compared with the sham group and the NK-92MI-GFP cell group (P < 0.05). Interestingly, no statistically significant delay of tumor growth was found with treatment of NK-92MI-GFP cells (Fig. 6A and B), even though NK-92MI-GFP cells selectively accumulated in tumor tissues (Fig. 6C). There was no difference between the tumor-infiltrating efficiency of NK-92MI-FETZ cells and that of NK-92MI-GFP cells (data not shown). Data from immunohistochemistry staining assay show the presence of both NK-92MI-GFP cells and NK-92MI-FETZ cells in tumor tissues (CD45 in Fig. 6D). However, NK-92MI-FETZ cells effectively induced apoptosis (TUNEL in Fig. 6D). Notably, ELISA data showed a 6-fold and 7-fold increase in TRAIL concentrations in the blood (Fig. 6E) and tumor tissues (Fig. 6F) of the NK-92MI-FETZ cell–treated group, respectively, compared with those of the sham group and the NK-92MI-GFP cell–treated group. Collectively, these results show that NK-92MI-FETZ cells can effectively accumulate at tumor sites, infiltrate into peritoneal tumors via i.p. injection, and induce apoptotic tumor cell death via the secretion of TRAIL in vivo.

Figure 6.

Effect of NK-92MI-FETZ on the growth of LS174T i.p. tumors. NOD/SCID mice were i.p. inoculated with 1 × 106 LS174T-luc+ cells. Four days after tumor inoculation, all tumor-bearing mice were treated with either PBS alone (sham), NK-92MI-GFP (5 × 106) cells, or NK-92MI-FETZ (5 × 106) cells by i.p. injection twice a week for 2 weeks. A, mice were imaged using the IVIS Imaging System Series 200 twice a week. Representative images are shown on day 25. B, line graph illustrating the luciferase activities (photons/second) in LS174T tumor-bearing mice, which were treated with PBS, NK-92MI-GFP, or NK-92MI-FETZ from day 4 to day 14 (arrows), were determined until day 25. *, P < 0.05, statistically significant difference compared with the sham group. #, P < 0.05, statistically significant difference compared with the NK-92MI-GFP group. C, after sacrifice of the mice, the liver, lung, and tumor tissue were harvested (three mice/group). The presence of green fluorescence in each group was detected with the IVIS system. Representative photos are shown. D, tumor tissues were harvested on day 28 and subjected to TUNEL assay, hematoxylin and eosin (H&E) staining, and immunohistochemistry staining with anti-CD45 primary antibodies. Cell nuclei were stained with DAPI. GFP (NK-92MI cell) fluorescent signals were detected. Arrows, CD45-positive NK-92MI cells. Representative images are shown (magnification, ×400). E and F, serum (E) and tumor tissues (F) from each mouse were collected and subjected to ELISA assay. TRAIL concentration was quantified.

Figure 6.

Effect of NK-92MI-FETZ on the growth of LS174T i.p. tumors. NOD/SCID mice were i.p. inoculated with 1 × 106 LS174T-luc+ cells. Four days after tumor inoculation, all tumor-bearing mice were treated with either PBS alone (sham), NK-92MI-GFP (5 × 106) cells, or NK-92MI-FETZ (5 × 106) cells by i.p. injection twice a week for 2 weeks. A, mice were imaged using the IVIS Imaging System Series 200 twice a week. Representative images are shown on day 25. B, line graph illustrating the luciferase activities (photons/second) in LS174T tumor-bearing mice, which were treated with PBS, NK-92MI-GFP, or NK-92MI-FETZ from day 4 to day 14 (arrows), were determined until day 25. *, P < 0.05, statistically significant difference compared with the sham group. #, P < 0.05, statistically significant difference compared with the NK-92MI-GFP group. C, after sacrifice of the mice, the liver, lung, and tumor tissue were harvested (three mice/group). The presence of green fluorescence in each group was detected with the IVIS system. Representative photos are shown. D, tumor tissues were harvested on day 28 and subjected to TUNEL assay, hematoxylin and eosin (H&E) staining, and immunohistochemistry staining with anti-CD45 primary antibodies. Cell nuclei were stained with DAPI. GFP (NK-92MI cell) fluorescent signals were detected. Arrows, CD45-positive NK-92MI cells. Representative images are shown (magnification, ×400). E and F, serum (E) and tumor tissues (F) from each mouse were collected and subjected to ELISA assay. TRAIL concentration was quantified.

Close modal

Dramatic clinical antitumor effects have been observed with activated NK cells in refractory leukemia, but not ovarian cancer (33). The delivery mode of NK cells may affect tumor cell killing and circumvent the lack of NK-cell expansion, especially for ovarian cancer. We established a mouse peritoneal carcinomatosis xenograft model via i.p. injection of LS174T cells and confirmed that i.p.-, but not i.v.-delivered, NK-92MI cells were able to effectively infiltrate LS174T peritoneal tumors (Fig. 5C and E). These results were consistent with previous observations: NK cells not only accumulate selectively at tumor sites independent of tumor antigen expression, but also penetrate deeply into tumor tissues (10–13). Upon activation, infiltrated NK cells proliferate and upregulate effector molecules, such as perforin, granzymes, FasL, TNFα, and TRAIL (24–27). However, we observed that the effect of NK-92MI-GFP cells was limited (Fig. 6A and B), although NK-92MI-GFP cells infiltrated into tumors (Fig. 6C and D), engrafted within the peritoneal cavity, and circulated (data not shown). One possible limitation of infiltrated NK cells is due to tumor microenvironments. Tumor cells may secrete tumor-derived exosomes containing IL10 cytokine, TGFβ growth factor, and miRNAs, which recruit immunosuppressive cells and impact the NK functions (34). The other possibility is the absence of danger signals (22). A failure of integration of signals for activation and inhibition may cause the impairment of NK-cell functions. Another possibility is an insufficient ability to kill cancer cells through Fas-, perforin-mediated apoptosis, or NK-cell exhaustion (35). Among these possibilities, at the present time, we can only speculate on the inactivation of NK cells, which results from the unbalance of signals produced by activating receptors and inhibitory receptors. It is possible that the PD-1/PD-L1 signaling axis modulates the tumoricidal efficacy of NK-92MI cells (21). This possibility needs to be investigated by blocking the PD-1/PD-L1 signaling axis using PD-1– and PD-L1–specific Abs.

A major hindrance in studying NK cells is the resistance of these cells to gene transfer, such as lipid-based transfection and electroporation, etc. Among various technologies available for gene transfer, lentiviral-mediated transduction has been successful in introducing genes into NK cells (36). We also successfully transduced Lenti-GFP and Lenti-FETZ vector into NK-92MI cells (Fig. 2). We confirmed the secreted TRAIL fusion protein (FETZ) with Western blot and ELISA assay (Fig. 2C and E). We also confirmed that the secreted TRAIL fusion protein is the N-linked glycoprotein (Fig. 2D). Glycosylation serves a variety of structural and functional roles in membrane and secreted proteins, including proper folding and enhanced protein stability, whereas unglycosylated protein degrades quickly (37). The glycosylated secretory TRAIL revealed cytotoxicity and tumoricidal activity (Figs. 3, 4, and 6)

Taken together, our findings show that NK-92MI-FETZ cells enhanced cell death and apoptosis in several colorectal cancer cell lines through secretion of glycosylated TRAIL fusion protein in vitro. NK-92MI-FETZ cells delivered i.p. can infiltrate colorectal peritoneal tumors, inhibit xenograft tumor growth, and increase apoptotic tumor cell death in vivo. Adoptive transfer of allogeneic NK-92 cells has been demonstrated to be a safe and potentially beneficial therapy with successful antitumor effects, receiving FDA approval for testing in cancer patients (38–40). We believe that the present findings will stimulate further clinical validation of i.p. delivery of genetically engineered NK-92MI-FETZ cell immunotherapy in colorectal peritoneal carcinomatosis patients.

No potential conflicts of interest were disclosed.

Conception and design: X. Song, W.T. Kwon, Y.J. Lee

Development of methodology: X. Song

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): X. Song, S.-H. Hong, W.T. Kwon, L.M. Bailey, P. Basse

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X. Song, W.T. Kwon, Y.T. Kwon

Writing, review, and/or revision of the manuscript: X. Song, D.L. Bartlett, Y.T. Kwon, Y.J. Lee

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y.T. Kwon

Study supervision: Y.T. Kwon, Y.J. Lee

The authors thank Christine Heiner (Department of Surgery, University of Pittsburgh) for her critical reading of the article.

This work was supported by NCI grant R01CA140554 (to Y.J. Lee). This project used the University of Pittsburgh Cancer Institute Core Facility, and all authors except Y.T. Kwon were supported in part by award P30CA047904.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Walczak
H
,
Miller
RE
,
Ariail
K
,
Gliniak
B
,
Griffith
TS
,
Kubin
M
, et al
Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo
.
Nat Med
1999
;
5
:
157
63
.
2.
Ling
J
,
Herbst
R
,
Mendelson
D
,
Eckhardt
S
,
O'Dwyer
P
,
Ebbinghaus
S
, et al
Apo2L/TRAIL pharmacokinetics in a phase 1a trial in advanced cancer and lymphoma
.
J Clin Oncol, ASCO Annual Meeting Proceedings (Post-Meeting Edition)
2006
;
24
:
18S
:
3047
.
3.
Kelley
SK
,
Harris
LA
,
Xie
D
,
Deforge
L
,
Totpal
K
,
Bussiere
J
, et al
Preclinical studies to predict the disposition of Apo2L/tumor necrosis factor-related apoptosis-inducing ligand in humans: characterization of in vivo efficacy, pharmacokinetics, and safety
.
J Pharmacol Exp Ther
2001
;
299
:
31
8
.
4.
Xin
Y
,
Tohnya
T
,
Herbst
R
,
Mendelson
D
,
Eckhardt
S
,
O'Dwyer
P
, et al
Population pharmacokinetic (PPK) analysis of recombinant human Apo2L/TRAIL (rhApo2L/TRAIL) in a phase 1a study in advanced cancer and lymphoma
.
J Clin Oncol
2008
;
26
:
2525
.
5.
Ichikawa
K
,
Liu
W
,
Zhao
L
,
Wang
Z
,
Liu
D
,
Ohtsuka
T
, et al
Tumoricidal activity of a novel anti-human DR5 monoclonal antibody without hepatocyte cytotoxicity
.
Nat Med
2001
;
7
:
954
60
.
6.
Yada
A
,
Yazawa
M
,
Ishida
S
,
Yoshida
H
,
Ichikawa
K
,
Kurakata
S
, et al
A novel humanized anti-human death receptor 5 antibody CS-1008 induces apoptosis in tumor cells without toxicity in hepatocytes
.
Ann Oncol
2008
;
19
:
1060
7
.
7.
Song
X
,
Kim
SY
,
Lee
YJ
. 
The role of Bcl-xL in synergistic induction of apoptosis by mapatumumab and oxaliplatin in combination with hyperthermia on human colon cancer
.
Mol Cancer Res
2012
;
10
:
1567
79
.
8.
Song
X
,
Kim
SY
,
Zhou
Z
,
Lagasse
E
,
Kwon
YT
,
Lee
YJ
. 
Hyperthermia enhances mapatumumab-induced apoptotic death through ubiquitin-mediated degradation of cellular FLIP(long) in human colon cancer cells
.
Cell Death Dis
2013
;
4
:
e577
.
9.
Wang
H
,
Davis
JS
,
Wu
X
. 
Immunoglobulin Fc domain fusion to TRAIL significantly prolongs its plasma half-life and enhances its antitumor activity
.
Mol Cancer Ther
2014
;
13
:
643
50
.
10.
Yang
Q
,
Hokland
ME
,
Bryant
JL
,
Zhang
Y
,
Nannmark
U
,
Watkins
SC
, et al
Tumor-localization by adoptively transferred, interleukin-2-activated NK cells leads to destruction of well-established lung metastases
.
Int J Cancer
2003
;
105
:
512
9
.
11.
Edsparr
K
,
Basse
PH
,
Goldfarb
RH
,
Albertsson
P
. 
Matrix metalloproteinases in cytotoxic lymphocytes impact on tumour infiltration and immunomodulation
.
Cancer Microenviron
2011
;
4
:
351
60
.
12.
Goding
SR
,
Yang
Q
,
Knudsen
KB
,
Potter
DM
,
Basse
PH
. 
Cytokine gene therapy using adenovirally transduced, tumor-seeking activated natural killer cells
.
Hum Gene Ther
2007
;
18
:
701
11
.
13.
Goding
S
,
Yang
Q
,
Mi
Z
,
Robbins
PD
,
Basse
PH
. 
Targeting of products of genes to tumor sites using adoptively transferred A-NK and T-LAK cells
.
Cancer Gene Ther
2007
;
14
:
441
50
.
14.
Herberman
RB
,
Holden
HT
. 
Natural cell-mediated immunity
.
Adv Cancer Res
1978
;
27
:
305
77
.
15.
Herberman
RB
,
Holden
HT
,
Ting
CC
,
Lavrin
DL
,
Kirchner
H
. 
Cell-mediated immunity to leukemia virus- and tumor-associated antigens in mice
.
Cancer Res
1976
;
36
:
615
21
.
16.
Oehler
JR
,
Herberman
RB
. 
Natural cell-mediated cytotoxicity in rats. III. Effects of immunopharmacologic treatments on natural reactivity and on reactivity augmented by polyinosinic-polycytidylic acid
.
Int J Cancer
1978
;
21
:
221
9
.
17.
Ucisik-Akkaya
E
,
Dorak
MT
. 
A study of natural killer cell lectin-like receptor K1 gene (KLRK1/NKG2D) region polymorphisms in a European population sample
.
Tissue Antigens
2009
;
73
:
177
83
.
18.
Sivori
S
,
Carlomagno
S
,
Pesce
S
,
Moretta
A
,
Vitale
M
,
Marcenaro
E
. 
TLR/NCR/KIR: Which one to use and when?
Front Immunol
2014
;
5
:
105
.
19.
Tu
MM
,
Mahmoud
AB
,
Wight
A
,
Mottashed
A
,
Belanger
S
,
Rahim
MM
, et al
Ly49 family receptors are required for cancer immunosurveillance mediated by natural killer cells
.
Cancer Res
2014
;
74
:
3684
94
.
20.
Theorell
J
,
Gustavsson
AL
,
Tesi
B
,
Sigmundsson
K
,
Ljunggren
HG
,
Lundback
T
, et al
Immunomodulatory activity of commonly used drugs on Fc-receptor-mediated human natural killer cell activation
.
Cancer Immunol Immunother
2014
;
63
:
627
41
.
21.
Benson
DM
 Jr.
,
Bakan
CE
,
Mishra
A
,
Hofmeister
CC
,
Efebera
Y
,
Becknell
B
, 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
:
2286
94
.
22.
Long
EO
,
Kim
HS
,
Liu
D
,
Peterson
ME
,
Rajagopalan
S
. 
Controlling natural killer cell responses: Integration of signals for activation and inhibition
.
Ann Rev Immunol
2013
;
31
:
227
58
.
23.
Walzer
T
,
Dalod
M
,
Robbins
SH
,
Zitvogel
L
,
Vivier
E
. 
Natural-killer cells and dendritic cells: "l'union fait la force"
.
Blood
2005
;
106
:
2252
8
.
24.
Vivier
E
,
Tomasello
E
,
Baratin
M
,
Walzer
T
,
Ugolini
S
. 
Functions of natural killer cells
.
Nat Immunol
2008
;
9
:
503
10
.
25.
Zamai
L
,
Ahmad
M
,
Bennett
IM
,
Azzoni
L
,
Alnemri
ES
,
Perussia
B
. 
Natural killer (NK) cell-mediated cytotoxicity: Differential use of TRAIL and Fas ligand by immature and mature primary human NK cells
.
J Exp Med
1998
;
188
:
2375
80
.
26.
Wu
J
,
Lanier
LL
. 
Natural killer cells and cancer
.
Adv Cancer Res
2003
;
90
:
127
56
.
27.
Yokoyama
WM
,
Kim
S
,
French
AR
. 
The dynamic life of natural killer cells
.
Ann Rev Immunol
2004
;
22
:
405
29
.
28.
Velthuis
JH
,
de Bont
HJ
,
Medema
JP
,
Kuppen
PJ
,
Mulder
GJ
,
Nagelkerke
JF
. 
Interleukin-2 activated NK cells do not use the CD95L- and TRAIL-pathways in the rapid induction of apoptosis of rat colon carcinoma CC531s cells
.
Immunobiol
2003
;
207
:
115
27
.
29.
Whiteside
TL
,
Herberman
RB
. 
The role of natural killer cells in immune surveillance of cancer
.
Curr Opin Immunol
1995
;
7
:
704
10
.
30.
Mace
EM
,
Monkley
SJ
,
Critchley
DR
,
Takei
F
. 
A dual role for talin in NK cell cytotoxicity: activation of LFA-1-mediated cell adhesion and polarization of NK cells
.
J Immunol
2009
;
182
:
948
56
.
31.
Mandal
A
,
Viswanathan
C
. 
Natural killer cells: In health and disease
.
Hematol Oncol Stem Cell Ther
2015
;
8
:
47
55
.
32.
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
:
652
8
.
33.
Geller
MA
,
Cooley
S
,
Judson
PL
,
Ghebre
R
,
Carson
LF
,
Argenta
PA
, et al
A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer
.
Cytotherapy
2011
;
13
:
98
107
.
34.
Baginska
J
,
Viry
E
,
Paggetti
J
,
Medves
S
,
Berchem
G
,
Moussay
E
, et al
The critical role of the tumor microenvironment in shaping natural killer cell-mediated anti-tumor immunity
.
Front Immunol
2013
;
4
:
490
.
35.
da Silva
IP
,
Gallois
A
,
Jimenez-Baranda
S
,
Khan
S
,
Anderson
AC
,
Kuchroo
VK
, et al
Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade
.
Cancer Immunol Res
2014
;
2
:
410
22
.
36.
Micucci
F
,
Zingoni
A
,
Piccoli
M
,
Frati
L
,
Santoni
A
,
Galandrini
R
. 
High-efficient lentiviral vector-mediated gene transfer into primary human NK cells
.
Exp Hematol
2006
;
34
:
1344
52
.
37.
Sarkar
A
,
Wintrode
PL
. 
Effects of glycosylation on the stability and flexibility of a metastable protein: The human serpin alpha(1)-antitrypsin
.
Int J Mass Spectrom
2011
;
302
:
69
75
.
38.
Tonn
T
,
Becker
S
,
Esser
R
,
Schwabe
D
,
Seifried
E
. 
Cellular immunotherapy of malignancies using the clonal natural killer cell line NK-92
.
J Hematother Stem Cell Res
2001
;
10
:
535
44
.
39.
Klingemann
HG
. 
Natural killer cell-based immunotherapeutic strategies
.
Cytotherapy
2005
;
7
:
16
22
.
40.
Malmberg
KJ
,
Bryceson
YT
,
Carlsten
M
,
Andersson
S
,
Bjorklund
A
,
Bjorkstrom
NK
et al 
NK cell-mediated targeting of human cancer and possibilities for new means of immunotherapy
.
Cancer Immunol Immunother
2008
;
57
:
1541
52
.