The administration of low dose interleukin-2 (IL-2) results in a selective expansion of natural killer (NK) cells in vivo, and promotes the differentiation of NK cells from hematopoietic precursor cells in vitro. We have previously shown that stem cell factor (SCF ), the ligand to the c-kit tyrosine kinase receptor, enhances IL-2–induced NK cell proliferation and differentiation in vitro. Here, we investigated the effects of SCF plus IL-2 delivered to mice in vivo. Eight-week-old C57BL/6 mice were treated with a continuous subcutaneous infusion of IL-2 (1 × 104 IU/d) plus a daily intraperitoneal dose of SCF (100 μg/kg/d), IL-2 alone, SCF alone, or vehicle alone for 8 weeks. The in vivo serum concentration of IL-2 ranged between 352 ± 12.0 pg/mL and 606 ± 9.0 pg/mL, achieving selective saturation of the high affinity IL-2 receptor, while the peak SCF serum concentration was 296 ± 13.09 ng/mL. Alone, the daily administration of SCF had no effect on the expansion of NK cells. The continuous infusion of IL-2 alone did result in a significant expansion of NK1.1+CD3 cells compared to mice treated with placebo or SCF. However, mice treated with both SCF and IL-2 showed an increase in the absolute number of NK cells that was more than twofold that seen with IL-2 alone, in the spleen (P ≤ .005), bone marrow (P ≤ .025), and blood (P < .05). NK cytotoxic activity against YAC-1 target cells was significantly higher for mice treated with SCF plus IL-2, compared to mice treated with IL-2 alone (P ≤ .0005). Interferon-γ (IFN-γ) production in cytokine-activated splenocytes was also greater for the SCF plus IL-2 group, over IL-2 treatment alone (P ≤ .01). The effect of SCF plus IL-2 on NK cell expansion was likely mediated via NK cell precursors, rather than mature NK cells. In summary, we provide the first evidence that SCF can significantly enhance expansion of functional NK cells induced by the prolonged administration of low dose IL-2 in vivo. Since the NK cell is a cytotoxic innate immune effector and a potent source of IFN-γ, this therapeutic strategy for NK cell expansion may serve to further enhance innate immune surveillance against malignant transformation and infection in the setting of cancer and/or immunodeficiency.

NATURAL KILLER (NK) cells are bone marrow (BM)-derived large granular lymphocytes that mediate non-major histocompatibility complex (MHC) restricted cytotoxicity, and are a critical component of the innate immune response to viral and obligate intracellular parasitic infection.1-4 Murine NK cells lack CD3 and can express the NK1.1 surface antigen, and are functionally characterized by their ability to spontaneously lyse tumor-derived target cells such as YAC-1.5 Murine6 and human7,8 NK cells constitutively express functional forms of the interleukin-2 receptor (IL-2R). The daily subcutaneous administration of low dose IL-2 can selectively expand NK cells in vivo in both mice9 and humans.10,11 Further, numerous studies have described the differentiation of NK cells from murine and human BM hematopoietic precursor cell (HPC) cultures with IL-2 in vitro.12-19 

The proto-oncogene c-kit encodes a tyrosine kinase receptor that is expressed on normal HPCs at various stages of maturation.20-23 Stem cell factor (SCF, kit ligand, or steel factor) is a ligand for the c-kit receptor.24-26 While SCF alone has little effect on HPC differentiation,27 it acts synergistically with numerous hematopoietic growth factors, such as IL-3, IL-6, erythropoietin (EPO), granulocyte colony-stimulating factor, and granulocyte-macrophage colony-stimulating factor (GM-CSF ), to augment their effects on HPC differentiation in vitro28-30 and in vivo.31-35 A subset of human NK cells has been shown to express the c-kit receptor, and exposure to SCF in vitro promotes their survival and potentiates their proliferative response to IL-2.36,37 Highly purified human CD34+ HPCs are induced to differentiate and expand (6- to 16-fold) into CD56+ NK cells in the presence of IL-2 plus SCF, while the absence of SCF led to a dramatic reduction in NK cell expansion without affecting differentiation.19 Collectively, these data suggested that SCF may enhance IL-2–induced NK cell expansion in vivo, and the current study was performed to test this hypothesis.

Animals. Six-week-old female C57BL/6 mice were obtained from Taconic Farms (Germantown, NY), and were housed in microisolator cages within a pathogen-free animal facility at the Roswell Park Cancer Institute. Mice were 8 weeks of age with an average weight of 23 g when treatment was initiated.

Cell lines. NK cytotoxicity assays were performed with the NK-sensitive target cell line YAC-1, grown in RPMI-1640 supplemented with 10% heat-inactivated fetal calf serum (FCS; Sigma, St Louis, MO), anti-PPLO agent, and antibiotics (GIBCO-BRL, Grand Island, NY).

Antibodies. Hamster anti-mouse CD3-fluorescein isothiocyanate (FITC), rat anti-mouse IL-2Rβ-FITC, rat anti-mouse c-kit-FITC, mouse anti-mouse NK1.1-phycoerythrin (PE) conjugated monoclonal antibodies (MoAbs), rat anti-mouse CD16/CD32 (Fc Block), and rat PE- and FITC-conjugated nonreactive mouse isotype control MoAb were purchased from Pharmingen (San Diego, CA). PE- and FITC-conjugated nonreactive mouse isotype control MoAbs were purchased from Becton Dickinson (San Jose, CA), Unlabeled nonreactive mouse Ig (IgG1 ) was obtained from Sigma, and hamster Ig (IgG1 ) was obtained from Pharmingen.

Cytokines. Recombinant-methionyl human (rh)IL-2 (specific activity 7.4 × 106 U/mg, <2.5 endotoxin units (EU) / mg protein) and polyethylene glycol (PEG)-complexed recombinant rat (rr)SCF (<1 EU / mg protein) were kindly provided by Amgen Inc (Thousand Oaks, CA). For in vitro stimulation of unfractionated splenocytes, rhIL-2 (Hoffman-LaRoche, Nutley, NJ, specific activity 1.5 × 107 U/mg) and murine rIL-12 (Genetics Institute, Cambridge, MA, specific activity 4.6 × 106 U/mg) were used. For in vivo pharmacokinetic (PK) studies rhSCF-PEG (Amgen) was used.

Drug administration. Four treatment groups of mice consisted of (1) placebo (vehicle only) (n = 5); (2) rrSCF plus vehicle (n = 6); (3) rhIL-2 plus vehicle (n = 6); (4) rhIL-2 plus rrSCF (n = 6), all for 8 weeks duration. IL-2 was administered via Alzet osmotic mini-pumps (Alza Corp, Palo Alto, CA), set to deliver 1 × 104 IU per day in a vehicle of phosphate-buffered saline (PBS; GIBCO) containing 0.1% syngeneic mouse serum (Taconic Farms). The osmotic pumps were inserted subcutaneously (s.c.) on the dorsum of the mice following the manufacturer's instructions and replaced every 10 days. Mice not treated with IL-2 were fitted with mini-pumps set to deliver an equal volume of vehicle as the IL-2–treated mice. SCF was injected intraperitoneally (i.p.) once daily at a dose of 100 μg/kg body weight in a volume of 100 μL PBS containing 0.1% syngeneic mouse serum. Mice not treated with SCF received a daily i.p. injection of vehicle only. All mice were therefore manipulated identically with regard to drug administration. PK for the administration of IL-2 via the s.c. pumps and SCF i.p. injection were performed by collecting serum at the indicated time points, and assayed by enzyme-linked immunosorbent assay (ELISA) for IL-2 (Endogen, Woburn, MA) or SCF (Amgen Inc). PK studies for SCF-PEG were performed with rhSCF-PEG so that exogenously administered SCF-PEG and endogenously produced mouse SCF could be distinguished. The ELISA used to measure rhSCF-PEG levels (Amgen) does not cross react with mouse SCF. Serum samples were not collected frequently enough to calculate area under the curve. All animal procedures were approved by the Institute Animal Care and Use Committee.

Isolation of spleen, BM, and blood cells. Mice were anesthetized with ether and killed by cervical dislocation. Blood was obtained by direct cardiac puncture. Spleens were excised, weighed, disrupted, and passed through stainless steel mesh into RPMI-1640 supplemented with 10% fetal calf serum (FCS). Splenocytes were then gently pipetted to create a single cell suspension. In some instances indicated below, splenocytes were then passed through a nylon wool column, and the nonadherent NK1.1+CD3 lymphocytes were sorted to >98% purity on a FACStar Plus flow cytometer (Becton Dickinson). BM from each mouse was obtained from two femurs by flushing the bone shaft with RPMI-1640 plus 10% FCS with a 27 gauge needle. Contaminating red blood cells (RBCs) were removed from the single cell suspensions of all tissues by incubation for 5 minutes at room temperature with sterile ammonium chloride/potassium lysis buffer. After a final resuspension in RPMI-1640 plus 10% FCS, the total number of cells from each tissue were enumerated with a hemocytometer.

Cytotoxicity assay. Unfractionated splenocytes were assessed for NK cytotoxic activity in a standard 51Cr release assay against YAC-1 tumor cells. One million YAC-1 target cells were labeled with 100 μCi of 51Cr for 1 hour, washed four times, and plated at 5 × 103 cells per well in a 96-well V-bottom plate. Splenocytes were freshly isolated from each mouse and incubated with YAC-1 target cells at an E/T ratio of 200:1 for 4 hours at 37°C in a total volume of 200 μL, in the absence of any exogenous cytokines. One hundred microliters of supernatant was then procured from each well and counted on a gamma counter to determine experimental release. Spontaneous release was determined from labeled target cells incubated in medium alone, while maximal release was obtained from labeled targets lysed in 1% NP-40. Percent specific cytotoxicity = [(experimental release − spontaneous release) / (maximal release − spontaneous release)] × 100.

Flow cytometric analysis. Approximately 1 × 106 spleen, BM, and blood cells were each incubated with nonreactive, unlabeled mouse and hamster Ig for 15 minutes on ice, and then stained with PE- and FITC-conjugated nonreactive isotype control MoAb, or anti-NK1.1-PE plus anti-CD3-FITC MoAb, for 30 minutes on ice. Cells were then washed twice with PBS and resuspended in 1 mL of 1% formalin in PBS, and forward scatter, side scatter, and fluorescence data were collected on 10,000 events using a FACScan (Becton Dickinson, Mountain View, CA). Flow cytometric data were then analyzed using the Winlist software program (Verity Software House, Topsham, ME), and results displayed in 2 parameter orthographic dot plots, with percentages indicating the fraction of total ungated events collected. Control PE- and FITC-stained cells were used to set quadrant gates, with >99% of control events designated as background and nonspecific fluorescence.

Analysis of cytokine production. 5 × 105 unfractionated splenocytes from each mouse were plated with 200 μL medium (RPMI-1640 plus 10% FCS), rhIL-2 (10 ng/mL) plus rmIL-12 (30 ng/mL), in 96-well U-bottom plates. Cell cultures were then incubated at 37°C for 72 hours, after which cell-free supernatants were harvested and assayed by ELISA (Endogen) for murine IFN-γ.

Proliferation and immunomodulation assays. Purified (>98%) NK1.1+CD3 splenocytes were plated in RPMI-1640 supplemented with 10% FCS, 5 μmol/L β-mercaptoethanol (BME) and antibiotics at a concentration of 1 × 105/well. Cells were cultured for 24 and 48 hours in one of four conditions: (1) medium alone; (2) rrSCF-PEG (100 ng/mL); (3) rhIL-2 (10 ng/mL); (4) SCF plus IL-2. Cells were then procured, preincubated with Fc Block (Pharmingen), and stained with anti-NK1.1-PE and either anti-c-kit-FITC, anti-IL-2Rβ-FITC, or isotype control-FITC MoAb. Ten thousand events were collected and analyzed as described above. For assessment of proliferation, 5 × 104 sorted NK1.1+CD3 cells were plated in triplicate in 200 μL of medium with an increasing concentration of rhIL-2 (0.001 nmol/L, 0.01 nmol/L, 0.1 nmol/L, 1 nmol/L, and 10 nmol/L) with and without 100 ng/mL rrSCF-PEG, for 72 hours at 37°C. During the final 24 hours of incubation, 3H-thymidine was added to the cultures. Cells were then procured and proliferation was assessed by measuring 3H-thymidine incorporation. A similar assay was performed for 7 days, at which time cells were counted using a hemocytometer and viability assessed by vital dye exclusion.

Statistical analysis. All comparisons between treatment groups were performed using the unpaired Student's t-test, with P < .05 designated as significant.

Pharmacokinetics of IL-2 and SCF administration in vivo. Eight-week-old female C57BL/6 mice were treated with a continuous s.c. infusion of IL-2 at a dose of 1 × 104 IU per day via osmotic mini-pumps, and/or SCF at a dose of 100 μg/kg body weight by once-daily i.p. injection, each for 8 weeks duration. PK of IL-2 delivered by this method resulted in serum concentrations in the range of 352 ± 12.0 pg/mL to 606 ± 9.0 pg/mL (21 to 88.5 picomolar). Thus, serum IL-2 concentrations were capable of saturating the high affinity IL-2Rαβγ, and partially saturating the intermediate affinity IL-2Rβγ (Fig 1A).38 A single i.p. injection of rSCF resulted in measurable serum concentrations over 24 hours, peaking at 296.93 ± 13.09 ng/mL (Fig 1B).

Fig. 1.

Pharmacokinetics of rhIL-2 and rhSCF-PEG treatment in mice. (A) 1 × 104 IU per day rIL-2 was delivered by constant subcutaneous infusion via Alzet osmotic pump, and measurement of serum IL-2 was performed by ELISA on days 0, 4, 10, and 12 after implantation. (B) PK of rhSCF-PEG administered as a single i.p. injection of 100 μg/kg body weight. Serum levels were measured by ELISA 0, 4, 8, 12, and 24 hours after injection. Results for each cytokine represent the mean ± SEM obtained from two mice with each measurement performed in duplicate.

Fig. 1.

Pharmacokinetics of rhIL-2 and rhSCF-PEG treatment in mice. (A) 1 × 104 IU per day rIL-2 was delivered by constant subcutaneous infusion via Alzet osmotic pump, and measurement of serum IL-2 was performed by ELISA on days 0, 4, 10, and 12 after implantation. (B) PK of rhSCF-PEG administered as a single i.p. injection of 100 μg/kg body weight. Serum levels were measured by ELISA 0, 4, 8, 12, and 24 hours after injection. Results for each cytokine represent the mean ± SEM obtained from two mice with each measurement performed in duplicate.

Close modal

SCF enhances IL-2–mediated expansion of murine NK cells in the spleen. Administration of SCF alone or IL-2 alone resulted in a significant increase in spleen weight more than that seen with placebo (P ≤ .005 and P ≤ .0005, respectively). However, the concomitant administration of SCF and IL-2 resulted in a further significant increase in spleen weight more than that seen with either factor alone (Fig 2A, IL-2 v IL-2 plus SCF, P < .05). Total splenic cell number was significantly increased with IL-2 treatment more than either placebo or SCF alone (P ≤ .0005), and the combination of SCF plus IL-2 resulted in a further significant increase in total cell number compared to IL-2 treatment alone (P ≤ .025, Fig 2B). Flow cytometric analysis of the spleen cell populations showed that SCF did not increase the absolute number of NK1.1+CD3 NK cells more than placebo, however, IL-2 did (P ≤ .005). Moreover, the combination of SCF and IL-2 increased the absolute number of splenic NK1.1+CD3 NK cells more than twofold above IL-2 treatment alone (P ≤ .005, Fig 2C). The percentage of splenic NK1.1+CD3 NK cells in mice treated with SCF plus IL-2 was also significantly increased more than that seen with IL-2 alone (P < .005). Thus, for each splenic index the combination of SCF and IL-2 treatment resulted in a significant increase more than that measured with each factor alone.

Fig. 2.

SCF enhances IL-2 mediated expansion of splenic NK cells. Four groups of C57BL/6 mice (n = 5 to 6 per group) were treated with placebo, SCF, IL-2, or a combination of SCF plus IL-2, as described in Materials and Methods. After 8 weeks, spleens were excised and weighed (A) and the absolute number of spleen cells were determined by cell enumeration with a hemocytometer (B). 1 × 106 spleen cells were stained with NK1.1-PE and CD3-FITC MoAb and analyzed by flow cytometry for the NK1.1+CD3 NK cell population. There were 2.9 ± 0.2% NK1.1+CD3 cells in the control group, 1.8 ± 0.3% in the SCF group, 1.8 ± 0.1% in the IL-2 group, and 3.2 ± 0.2% in the SCF plus IL-2 group. The SCF plus IL-2 group had a significantly greater increase in percent NK1.1+CD3 cells compared to the IL-2 alone group (P < .005). Absolute splenic NK cell number was then calculated by multiplying the percentage NK1.1+CD3 NK cells by the total spleen cell number (C). The results represent the mean weights, total splenic cell number, and absolute splenic NK cell number of each treatment group ± SEM. Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group are indicated by the asterisk, and represent P < .05 (A), P ≤ .025 (B) and P ≤ .005 (C).

Fig. 2.

SCF enhances IL-2 mediated expansion of splenic NK cells. Four groups of C57BL/6 mice (n = 5 to 6 per group) were treated with placebo, SCF, IL-2, or a combination of SCF plus IL-2, as described in Materials and Methods. After 8 weeks, spleens were excised and weighed (A) and the absolute number of spleen cells were determined by cell enumeration with a hemocytometer (B). 1 × 106 spleen cells were stained with NK1.1-PE and CD3-FITC MoAb and analyzed by flow cytometry for the NK1.1+CD3 NK cell population. There were 2.9 ± 0.2% NK1.1+CD3 cells in the control group, 1.8 ± 0.3% in the SCF group, 1.8 ± 0.1% in the IL-2 group, and 3.2 ± 0.2% in the SCF plus IL-2 group. The SCF plus IL-2 group had a significantly greater increase in percent NK1.1+CD3 cells compared to the IL-2 alone group (P < .005). Absolute splenic NK cell number was then calculated by multiplying the percentage NK1.1+CD3 NK cells by the total spleen cell number (C). The results represent the mean weights, total splenic cell number, and absolute splenic NK cell number of each treatment group ± SEM. Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group are indicated by the asterisk, and represent P < .05 (A), P ≤ .025 (B) and P ≤ .005 (C).

Close modal

SCF enhances IL-2–mediated expansion of murine NK cells in BM and blood. Treatment with SCF alone did not alter the total number of BM cells or the absolute number of BM NK1.1+CD3 NK cells compared to placebo-treated controls. Treatment with IL-2 alone significantly increased both the total cell number (P ≤ .025) and the absolute number of NK cells (P ≤ .005) in BM over placebo and SCF alone. The total number of BM cells significantly increased even further when SCF was combined with IL-2 (P < .05, Fig 3A), and the absolute number of NK1.1+CD3 NK cells doubled with the administration of SCF and IL-2, compared to IL-2 treatment alone (P ≤ .025, Fig 3B). The percentage of BM NK1.1+CD3 NK cells in mice treated with SCF plus IL-2 was also significantly increased over that seen with IL-2 alone (P < .005). Likewise, total blood leukocyte number and absolute NK cell number did not increase with SCF alone, but were significantly increased in IL-2 treated mice over both placebo- and SCF-treated mice (P ≤ .025 and P ≤ .005, respectively, Fig 4). The combination of SCF and IL-2 treatment further increased both the total blood leukocyte number and absolute NK cell number over that seen with IL-2 treatment alone (P < .05, Fig 4). However, the percentage of blood NK1.1+CD3 NK cells in mice treated with SCF plus IL-2 was not significantly increased over that seen with IL-2 alone.

Fig. 3.

SCF enhances IL-2 mediated expansion of BM NK cells. After eight weeks of treatment, BM was procured from two femurs in each mouse and the absolute number of BM cells were determined by cell enumeration with a hemocytometer (A). 1 × 106 BM cells were then stained with NK1.1-PE and CD3-FITC and analyzed by flow cytometry for NK1.1+CD3 NK cell populations. There were 1.3 ± 0.02% NK1.1+CD3 cells in the control group, 1.4 ± 0.2% in the SCF group, 1.7 ± 0.1% in the IL-2 group, and 2.4 ± 0.2% in the SCF plus IL-2 group. The SCF plus IL-2 group had a significantly greater increase in percent NK1.1+CD3 cells compared to the IL-2 alone group (P < .005). Absolute BM NK cell number was calculated by multiplying total BM cells by the percent NK cells in the BM (B). Results represent the mean absolute number from 5 to 6 mice per treatment group ± SEM. Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group is indicated by the asterisk, and represent P < .05 (A), and P ≤ .025 (B).

Fig. 3.

SCF enhances IL-2 mediated expansion of BM NK cells. After eight weeks of treatment, BM was procured from two femurs in each mouse and the absolute number of BM cells were determined by cell enumeration with a hemocytometer (A). 1 × 106 BM cells were then stained with NK1.1-PE and CD3-FITC and analyzed by flow cytometry for NK1.1+CD3 NK cell populations. There were 1.3 ± 0.02% NK1.1+CD3 cells in the control group, 1.4 ± 0.2% in the SCF group, 1.7 ± 0.1% in the IL-2 group, and 2.4 ± 0.2% in the SCF plus IL-2 group. The SCF plus IL-2 group had a significantly greater increase in percent NK1.1+CD3 cells compared to the IL-2 alone group (P < .005). Absolute BM NK cell number was calculated by multiplying total BM cells by the percent NK cells in the BM (B). Results represent the mean absolute number from 5 to 6 mice per treatment group ± SEM. Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group is indicated by the asterisk, and represent P < .05 (A), and P ≤ .025 (B).

Close modal
Fig. 4.

SCF enhances IL-2–mediated expansion of NK cells in the blood. After eight weeks of treatment absolute leukocyte number per mL in blood was obtained (A) and then multiplied by the percent NK1.1+CD3 NK cells determined by flow cytometry to yield the absolute number of NK cells per mL of blood (B). There were 4.8 ± 0.8% NK1.1+CD3 cells in the control group, 4.5 ± 0.3 in the SCF group, 10.3 ± 2.8% in the IL-2 group, and 9.8 ± 1.1% in the SCF plus IL-2 group. Results represent the mean absolute number from 5 to 6 mice in each treatment group ± SEM. Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group are indicated by the asterisk, and represent P < .05 (A and B).

Fig. 4.

SCF enhances IL-2–mediated expansion of NK cells in the blood. After eight weeks of treatment absolute leukocyte number per mL in blood was obtained (A) and then multiplied by the percent NK1.1+CD3 NK cells determined by flow cytometry to yield the absolute number of NK cells per mL of blood (B). There were 4.8 ± 0.8% NK1.1+CD3 cells in the control group, 4.5 ± 0.3 in the SCF group, 10.3 ± 2.8% in the IL-2 group, and 9.8 ± 1.1% in the SCF plus IL-2 group. Results represent the mean absolute number from 5 to 6 mice in each treatment group ± SEM. Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group are indicated by the asterisk, and represent P < .05 (A and B).

Close modal

SCF augments IL-2–mediated increases in NK cell function. After observing the increase in NK1.1+CD3 NK cells, we sought to confirm those data by assessing NK cell function. When equal numbers of splenocytes were obtained from mice treated with placebo, SCF, IL-2, or SCF plus IL-2, and assayed for their ability to lyse YAC-1 target cells in the absence of any exogenous cytokines, cytotoxicity was greatest with splenocytes obtained from mice treated with SCF plus IL-2 (IL-2 v IL-2 plus SCF, P ≤ .0005, Fig 5A). In a second assay of NK cell function, equal numbers of unfractionated splenocytes from each of the four treatment groups were plated and stimulated in vitro with a combination of rhIL-2 (10 ng/mL) and rmIL-12 (30 ng/mL) for 72 hours, a known costimulus for NK IFN-γ production.39 IFN-γ production was significantly greater in splenocytes obtained from mice treated with SCF plus IL-2, compared with all other groups (P ≤ .01, Fig 5B). Collectively, these functional data support the phenotypic data showing a significant expansion of NK1.1+CD3 NK cells in mice treated with the combination of SCF plus IL-2 over that seen with placebo or either cytokine alone.

Fig. 5.

Functional assessment of NK cell expansion. Fresh single cell suspensions of unfractionated splenocytes were obtained from mice in each of the indicated treatment groups (n = 5 to 6 per group) and then assayed for NK cytotoxicity against YAC-1 targets (200:1 E/T ratio) in a standard 4 hour 51Cr release assay, without the addition of exogenous cytokines (A). Unfractionated splenocytes were also cultured (2 × 105 cells per well) in rhIL-2 plus rmIL-12 for 72 hours, after which cell-free culture supernatants were harvested and assayed for murine IFN-γ by ELISA (B). Results represent the mean percent cytotoxicity (A) or IFN-γ production (B) ± SEM. For both assays, the percentage NK1.1+CD3 cells were 2.9 ± 0.2% NK1.1+CD3 cells in the control group, 1.8 ± 0.3% in the SCF group, 1.8 ± 0.1% in the IL-2 group, and 3.2 ± 0.2% in the SCF plus IL-2 group. The SCF plus IL-2 group had a significantly greater increase in percent NK1.1+CD3 cells compared to the IL-2 alone group (P < .005). Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group are indicated by the asterisk, and represent P ≤ .0005 (A), and P ≤ .01 (B).

Fig. 5.

Functional assessment of NK cell expansion. Fresh single cell suspensions of unfractionated splenocytes were obtained from mice in each of the indicated treatment groups (n = 5 to 6 per group) and then assayed for NK cytotoxicity against YAC-1 targets (200:1 E/T ratio) in a standard 4 hour 51Cr release assay, without the addition of exogenous cytokines (A). Unfractionated splenocytes were also cultured (2 × 105 cells per well) in rhIL-2 plus rmIL-12 for 72 hours, after which cell-free culture supernatants were harvested and assayed for murine IFN-γ by ELISA (B). Results represent the mean percent cytotoxicity (A) or IFN-γ production (B) ± SEM. For both assays, the percentage NK1.1+CD3 cells were 2.9 ± 0.2% NK1.1+CD3 cells in the control group, 1.8 ± 0.3% in the SCF group, 1.8 ± 0.1% in the IL-2 group, and 3.2 ± 0.2% in the SCF plus IL-2 group. The SCF plus IL-2 group had a significantly greater increase in percent NK1.1+CD3 cells compared to the IL-2 alone group (P < .005). Statistical significance comparing the SCF plus IL-2 group to the IL-2 alone group are indicated by the asterisk, and represent P ≤ .0005 (A), and P ≤ .01 (B).

Close modal

In vitro effects of SCF and/or IL-2 on mature NK1.1+CD3 splenocytes. To better understand the mechanism of in vivo NK cell expansion, we assessed mature NK cell expression of c-kit and IL-2Rβ, NK cell survival, and NK cell proliferation in the presence of SCF, IL-2, or SCF plus IL-2. On fresh NK1.1+CD3 splenocytes, 13.2 ± 0.27% coexpressed c-kit, and 89.04 ± 0.4% coexpressed IL-2Rβ. Cells cultured for 24 and 48 hours under conditions described above did not increase c-kit or IL-2Rβ expression on NK1.1+ cells (data not shown). Further, neither proliferation or survival of NK1.1+ cells were enhanced when cultured for 3 days or 7 days, respectively, in concentrations of IL-2 that were comparable to those obtained in vivo (ie, 0.01 to 0.1 nmol/L). The addition of rrSCF to these cultures had no effect on any of these parameters (data not shown). Hence, the effects of SCF and IL-2 on mouse NK cells in vivo are not likely to be predominantly mediated by expansion at the level of the mature NK1.1 cell compartment.

This report provides the first in vivo evidence that SCF can augment IL-2–induced expansion of murine NK cells. Significant expansion in absolute NK cell number over that seen with IL-2 alone was observed after 8 weeks of treatment with SCF plus IL-2 in all three hematopoietic compartments examined (ie, the spleen, BM, and blood). Therapy with SCF alone resulted in a modest increase in spleen weight, but did not result in any significant increase in total cell number or NK cell number in any tissue examined, compared to placebo-treated mice. Therefore, SCF itself does not appear to induce NK cell differentiation or expansion of NK cells in this system, but rather serves to enhance NK cell differentiation and expansion induced by IL-2.

To further characterize these measured increases in NK cell number, we assessed NK cell effector function in unfractionated splenocytes obtained from mice treated with placebo, SCF, IL-2, or the combination of SCF and IL-2. Cytotoxicity against YAC-1 target cells and early IFN-γ production following cytokine costimulation are both properties of murine NK cells.4,39 These analyses showed significantly greater NK effector function in unfractionated splenocytes obtained from mice treated with SCF plus IL-2 versus mice treated with IL-2 alone. These data are consistent with the significant increase in the number and percentage of NK1.1+CD3 spleen cells observed in mice treated with the combination of SCF plus IL-2 over those treated with IL-2 alone.

SCF combines with a number of cytokine receptor super family ligands to enhance the production of erythrocytes and leukocytes.27-30,35 SCF's synergy with EPO was recently shown to result from an activated c-kit–induced tyrosine phosphorylation of the EPO receptor's cytoplasmic domain not used by JAK2.40 SCF's synergy with GM-CSF in the human MO7e cell line occurs through increases in the cyclin dependent kinase inhibitor (cki) p21cip-1 cellular protein, and decreases in the cki p27kip-1.41 This alteration in the cki ratio was found to be correlated with increased cyclin dependent kinase 2 (cdk2) kinase activity, phosphorylation of the retinoblastoma (Rb) gene product, and ultimately enhanced proliferation. The mechanism by which activation of the c-kit tyrosine kinase receptor may enhance the signal induced by IL-2 binding to the IL-2R in NK cells or HPCs is currently unknown. The IL-2Rβ and IL-2Rγ are signal transducing subunits and members of the cytokine receptor super family. Whether mechanisms described above may be operative in the synergy observed between c-kit and IL-2Rβγ is not known.

The point of action of exogenously administered IL-2 and SCF in the NK cell differentiation schema is not known, but several possible scenarios exist. Recent studies performed on mice with 17β-estradiol-induced BM ablation showed that short-term (3 day) in vitro culture with IL-2 was able to substitute for the BM microenvironment, allowing maturation of functional NK1.1+CD3 NK cells.42 Mice with 17β-estradiol-induced BM ablation have functionally immature (nonlytic) NK cells in the spleen, and the authors showed that activation of the IL-2R supports maturation into lytic NK cell effectors. This is consistent with our observations that IL-2 alone expands functional NK cells in mice. Additionally, Aiba et al43 have recently examined the cytokine requirements for maturation of murine NK cell progenitors from the fetal thymus into NK cell colonies in a cell-free culture system. These studies revealed that SCF was indispensable for formation of immature NK cell colonies, and both IL-2 and IL-15 increase the frequency of these colonies. Based on these cytokine requirements, one possible point of action of exogenously administered SCF and IL-2 is at the point of lineage commitment to NK cell development.

Our previous in vitro work has explored IL-2- and SCF-induced NK cell production from purified human CD34+ HPCs in humans. Twenty-one day culture in low dose IL-2 alone resulted in CD56+ NK cell differentiation with little NK cell expansion, while culture in SCF alone resulted in a modest expansion (5- to 13-fold) with no NK cell differentiation. When human HPCs were cultured in both IL-2 and SCF, we observed NK cell differentiation with considerably greater NK cell expansion (6- to 16-fold) compared to that seen with IL-2 alone.19 Identical results were obtained in vitro with CD34+ HPCs, SCF and IL-15, a cytokine that also uses the β and γ components of the IL-2R for binding and signal transduction.30 Collectively these in vitro human data also suggest that IL-2 and SCF can synergize for NK cell production at the level of an NK precursor cell. We have shown that at concentrations obtained in vivo, combinations of SCF and IL-2 have essentially no effect on survival or proliferation of mature mouse NK1.1+ cells, further supporting the notion that this effect is mediated via differentiation and expansion of HPCs. Indeed, other investigators have recently shown that c-kit+Sca-2+NK1.1 mouse HPCs can differentiate and expand into NK1.1+ NK cells in the presence of SCF, Flt3 ligand, IL-6, IL-7, and IL-15, the latter of which signals through components of the IL-2R (V. Kumar, personal communication, May 1997).

Low dose IL-2 has been administered by daily s.c. injection to patients with cancer and immunodeficiency for 8 to 12 weeks. The therapy produces low (ie, picomolar) concentrations of IL-2 in vivo, and consistently results in selective expansion of NK cells.10,11 The in vivo data presented in this report would suggest that the combination of SCF plus IL-2 might provide a greater and more consistent expansion of these innate immune effector cells than low dose IL-2 alone. Given their cytolytic properties and potent IFN-γ production, such NK cell expansion may be beneficial in patients with cancer and/or immunodeficiency.

We thank Dr Keith Langley at Amgen, Inc for performing the ELISA quantitation of serum rhSCF-PEG levels, and Vinay Kumar for critical review of the manuscript.

Supported by National Institutes of Health (Bethesda, MD) Grants CA68326, CA65670, and CA68458. T.A.F. is the recipient of the Howard Hughes Medical Institute (Chevy Chase, MD) Research Fellowship for Medical Students. W.E.C. is the recipient of the American Society of Clinical Oncology (Alexandria, VA) Young Investigator Award.

Address reprint requests to Michael A. Caligiuri, MD, The Ohio State University, Arthur G. James Cancer Center, 300 W 10th Ave, Columbus, OH 43210.

1
Robertson
MJ
Ritz
J
Biology and clinical relevance of human natural killer cells.
Blood
76
1990
2421
2
Trinchieri
G
Biology of natural killer cells.
Adv Immunol
47
1989
187
3
Bancroft
GJ
The role of natural killer cells in innate resistance to infection.
Curr Opin Immunol
5
1993
503
4
Biron
CA
Gazzinelli
RT
Effects of IL-12 on immune responses to microbial infections: A key mediator in regulating disease outcome.
Curr Opin Immunol
7
1995
485
5
Hackett
J Jr
Tutt
M
Lipscomb
M
Bennett
M
Koo
G
Kumar
V
Origin and differentiation of natural killer cells. II. Functional and morphologic studies of purified NK-1.1+ cells.
J Immunol
136
1986
3124
6
Tanaka
T
Tsudo
M
Karasuyama
H
Kitamura
F
Kono
T
Hatakeyama
M
Taniguchi
T
Miyasaka
M
A novel monoclonal antibody against murine IL-2 receptor beta-chain. Characterization of receptor expression in normal lymphoid cells and EL-4 cells.
J Immunol
147
1991
2222
7
Voss
SD
Sondel
PM
Robb
RJ
Characterization of the interleukin 2 receptors (IL-2R) expressed on human natural killer cells activated in vivo by IL-2: Association of the p64 IL-2R gamma chain with the IL-2R beta chain in functional intermediate-affinity IL-2R.
J Exp Med
176
1992
531
8
Caligiuri
MA
Zmuidzinas
A
Manley
TJ
Levine
H
Smith
KA
Ritz
J
Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors.
J Exp Med
171
1990
1509
9
Piguet
PF
Grau
G
Irle
C
Vassalli
P
Administration of recombinant interleukin 2 to mice enhances production of hemopoietic and natural killer cells.
Eur J Immunol
16
1986
1257
10
Bernstein
ZP
Porter
MM
Gould
M
Lipman
B
Bluman
EM
Stewart
CC
Hewitt
RG
Fyfe
G
Poiesz
B
Caligiuri
MA
Prolonged administration of low-dose interleukin-2 in human immunodeficiency virus-associated malignancy results in selective expansion of innate immune effectors without significant clinical toxicity.
Blood
86
1995
3287
11
Meropol
NJ
Porter
M
Blumenson
LE
Lindemann
MJ
P. PR
Vaickus
L
Loewen
GM
Creaven
PJ
Wilkes
KA
Giedlin
MA
Caligiuri
MA
Daily subcutaneous injection of low-dose interleukin 2 expands natural killer cells in vivo without significant toxicity.
Clin Cancer Res
2
1996
669
12
Lotzova
E
Savary
CA
Human natural killer cell development from bone marrow progenitors: Analysis of phenotype, cytotoxicity and growth.
Nat Immunity
12
1993
209
13
Lotzova
E
Savary
CA
Champlin
RE
Genesis of human oncolytic natural killer cells from primitive CD34+CD33− bone marrow progenitors.
J Immunol
150
1993
5263
14
Miller
JS
Verfaillie
C
McGlave
P
The generation of human natural killer cells from CD34+/DR− primitive progenitors in long-term bone marrow culture.
Blood
80
1992
2182
15
Miller
JS
Alley
KA
McGlave
P
Differentiation of natural killer (NK) cells from human primitive marrow progenitors in a stroma-based long-term culture system: Identification of a CD34+7+ NK progenitor.
Blood
83
1994
2594
16
Silva
MR
Hoffman
R
Srour
EF
Ascensao
JL
Generation of human natural killer cells from immature progenitors does not require marrow stromal cells.
Blood
84
1994
841
17
Shibuya
A
Nagayoshi
K
Nakamura
K
Nakauchi
H
Lymphokine requirement for the generation of natural killer cells from CD34+ hematopoietic progenitor cells.
Blood
85
1995
3538
18
Toksoz
D
Zsebo
KM
Smith
KA
Hu
S
Brankow
D
Suggs
SV
Martin
FH
Williams
DA
Support of human hematopoiesis in long-term bone marrow cultures by murine stromal cells selectively expressing the membrane-bound and secreted forms of the human homolog of the steel gene product, stem cell factor.
Proc Natl Acad Sci USA
89
1992
7350
19
Krajewski CA, Mrozek E, Caligiuri MA: Generation and expansion of CD3-CD56+ non-MHC-restricted cytotoxic effectors from human hematopoietic precursors: Requirement for c-kit ligand and interleukin 2. Proc Am Soc Clin Oncol 13:294, 1994 (abstr)
20
Yarden
Y
Kuang
WJ
Yang-Feng
T
Coussens
L
Munemitsu
S
Dull
TJ
Chen
E
Schlessinger
J
Francke
U
Ullrich
A
Human proto-oncogene c-kit: A new cell surface receptor tyrosine kinase for an unidentified ligand.
EMBO J
6
1987
3341
21
Chabot
B
Stephenson
DA
Chapman
VM
Besmer
P
Bernstein
A
The proto-oncogene c-kit encoding a transmembrane tyrosine kinase receptor maps to the mouse W locus.
Nature
335
1988
88
22
Geissler
EN
Ryan
MA
Housman
DE
The dominant-white spotting (W) locus of the mouse encodes the c-kit proto-oncogene.
Cell
55
1988
185
23
Ashman
LK
Cambareri
L
Bik
To L
Levinsky
RJ
Juttner
CA
Human proto-oncogene c-kit: A new cell surface receptor tyrosine kinase for an unidentified ligand.
Blood
78
1991
30
24
Williams
DE
Eisenman
J
Baird
A
Rauch
C
Van Ness
K
March
CJ
Park
LS
Martin
U
Mochizuki
DY
Boswell
HS
Burgess
GS
Cosman
D
Lyman
SD
Identification of a ligand for the c-kit proto-oncogene.
Cell
63
1990
167
25
Zsebo
KM
Wypych
J
McNiece
IK
Lu
HS
Smith
KA
Karkare
SB
Sachdev
RK
Yuschenkoff
VN
Birkett
NC
Williams
LR
Satyagal
VN
Tung
W
Bosselman
RA
Mediaz
EA
Langley
KE
Identification, purification, and biological characterization of hematopoietic stem cell factor from buffalo rat liver–conditioned medium.
Cell
63
1990
195
26
Huang
E
Nocka
K
Beier
DR
Chu
TY
Buck
J
Lahm
HW
Wellner
D
Leder
P
Besmer
P
The hematopoietic growth factor KL is encoded by the Sl locus and is the ligand of the c-kit receptor, the gene product of the W locus.
Cell
63
1990
225
27
de Vries
P
Brasel
KA
Eisenman
JR
Alpert
AR
Williams
DE
The effect of recombinant mast cell growth factor on purified murine hematopoietic stem cells.
J Exp Med
173
1991
1205
28
McNiece
IK
Langley
KE
Zsebo
KM
Recombinant human stem cell factor synergises with GM-CSF, G-CSF, IL-3 and epo to stimulate human progenitor cells of the myeloid and erythroid lineages.
Exp Hematol
19
1991
226
29
Avraham
H
Vannier
E
Cowley
S
Jiang
SX
Chi
S
Dinarello
CA
Zsebo
KM
Groopman
JE
Effects of the stem cell factor, c-kit ligand, on human megakaryocytic cells.
Blood
79
1992
365
30
Mrozek
E
Anderson
P
Caligiuri
MA
Role of interleukin-15 in the development of human CD56+ natural killer cells from CD34+ hematopoietic progenitor cells.
Blood
87
1996
2632
31
Andrews
RG
Briddell
RA
Knitter
GH
Opie
T
Bronsden
M
Myerson
D
Appelbaum
FR
McNiece
IK
In vivo synergy between recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in baboons enhanced circulation of progenitor cells.
Blood
84
1994
800
32
Andrews
RG
Briddell
RA
Knitter
GH
Rowley
SD
Appelbaum
FR
McNiece
IK
Rapid engraftment by peripheral blood progenitor cells mobilized by recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in nonhuman primates.
Blood
85
1995
15
33
Bodine
DM
Seidel
NE
Orlic
D
Bone marrow collected 14 days after in vivo administration of granulocyte colony-stimulating factor and stem cell factor to mice has 10-fold more repopulating ability than untreated bone marrow.
Blood
88
1996
89
34
de Haan
G
Donte
B
Engel
C
Loeffler
M
Nijhof
W
Prophylactic pretreatment of mice with hematopoietic growth factors induces expansion of primitive cell compartments and results in protection against 5-fluorouracil-induced toxicity.
Blood
87
1996
4581
35
Molineux
G
Migdalska
A
Szmitkowski
M
Zsebo
K
Dexter
TM
The effects on hematopoiesis of recombinant stem cell factor (ligand for c-kit) administered in vivo to mice either alone or in combination with granulocyte colony-stimulating factor.
Blood
78
1991
961
36
Matos
ME
Schnier
GS
Beecher
MS
Ashman
LK
William
DE
Caligiuri
MA
Expression of a functional c-kit receptor on a subset of natural killer cells.
J Exp Med
178
1993
1079
37
Carson
WE
Haldar
S
Baiocchi
RA
Croce
CM
Caligiuri
MA
The c-kit ligand suppresses apoptosis of human natural killer cells through the upregulation of bcl-2.
Proc Natl Acad Sci USA
91
1994
7553
38
Smith
KA
Interleukin-2: Inception, impact, and implications.
Science
240
1988
1169
39
Tripp
CS
Wolf
SF
Unanue
ER
Interleukin 12 and tumor necrosis factor alpha are costimulators of interferon gamma production by natural killer cells in severe combined immunodeficiency mice with listeriosis, and interleukin 10 is a physiologic antagonist.
Proc Natl Acad Sci USA
90
1993
3725
40
Wu
H
Klingmuller
U
Besmer
P
Lodish
HF
Interaction of the erythropoietin and stem-cell-factor receptors.
Nature
377
1995
242
41
Mantel
C
Luo
ZM
Canfield
J
Braun
S
Deng
CX
Broxmeyer
HE
Involvement of p21(cip-1) and p27(kip-1) in the molecular mechanisms of steel factor-induced proliferative synergy in vitro and of p21(cip-1) in the maintenance of stem/progenitor cells in vivo.
Blood
88
1996
3710
42
Puzanov
IJ
Bennett
M
Kumar
V
IL-15 can substitute for the marrow microenvironment in the differentiation of natural killer cells.
J Immunol
157
1996
4282
43
Aiba
Y
Hirayama
F
Ogawa
M
Clonal proliferation and cytokine requirement of murine progenitors for natural killer (NK) cells.
Blood
89
1997
4005
Sign in via your Institution