Open Access

Cytokine‑induced killer cells/natural killer cells combined with anti‑GD2 monoclonal antibody increase cell death rate in neuroblastoma SK‑N‑SH cells

  • Authors:
    • Chi Zhang
    • Xilin Xiong
    • Yang Li
    • Ke Huang
    • Ling Liu
    • Xiaomin Peng
    • Wenjun Weng
  • View Affiliations

  • Published online on: October 29, 2019     https://doi.org/10.3892/ol.2019.11020
  • Pages: 6525-6535
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neuroblastoma (NB) is one of the most common extracranial, solid, pediatric malignancies. Despite improvements in conventional therapies, including surgery, chemotherapy and radiation therapy, the prognosis of stage IV NB remains poor, indicating that novel treatment strategies are required. Immunotherapies, such as anti‑GD2 monoclonal antibodies, used alone or in combination with cytokines, and peripheral blood mononuclear cells or cord blood mononuclear cells (CBMNCs), have been indicated to cause NB cell death and to prolong patient survival in high‑risk NB; however, they remain limited by severe cytotoxicity and side effects. In the present study, it was determined that anti‑GD2 monoclonal antibody alone or CBMNC‑isolated cytokine‑induced killer (CIK)/natural killer (NK) cells alone significantly induced cell death of NB SK‑N‑SH cells, and the combination of anti‑GD2 antibody and CIK/NK cells could significantly increase the cell death rate compared with either treatment alone. In addition, based on a method referred to our previous study, it was identified that a two‑cytokine culture system, using interleukin IL‑2 and IL‑7, effectively stimulated the proliferation of CIK/NK cells. These results serve to suggest a novel treatment strategy for relapsed/refractory NB with high efficiency and few side effects.

Introduction

Neuroblastoma (NB) is one of the most common extracranial, solid, pediatric malignancies. NB originates from neural crest cells that can be destined for anywhere in the sympathetic nervous system (1), and accounts for 8–10% of pediatric malignancies and 15% of pediatric cancer-associated mortalities worldwide (2). NB is an extremely heterogeneous disease; for low-risk or intermediate-risk NB, the prognosis is favorable after surgical resection alone or with minimal chemotherapy (3). However, >60% of high-risk patients with NB die following recurrence, despite responding well to aggressive chemotherapy at the beginning of treatment (4). This suggests there is a lack of effective salvage regimens for patients with disease-recurrence.

Although the majority of patients with NB achieve complete remission under conventional treatments, including surgery, chemotherapy and radiotherapy, a number of patients still relapse in the late stages of chemotherapy, with acquired multi-drug resistance (MDR) (5). MDR contributes heavily to minimal residual disease (MRD) after chemotherapy, which leads to the recurrence of high-risk NB (6). Despite a large number of innovative studies on MRD, the prognosis of high-risk NB has remained poor over the past decade (79). Due to the issues with traditional chemotherapy drugs in NB, including increases in genetic mutations, dysregulated activity of tumor suppressor genes and serious side effects on organs (1012), the aim of the present study was to identify effective and well-tolerated methods for treating NB.

Ganglioside GD2 is a type of glycosphingolipid molecule that is uniformly expressed on the membrane of neurogenic tumor cells (13). The high expression level of GD2 in NB cells and its restricted distribution in normal tissues indicates that anti-GD2 monoclonal antibodies may be suitable for immunotherapy (14). Four types of anti-GD2 monoclonal antibody have been used in clinical trials, including murine anti-GD2 monoclonal antibody 3F8 (15), chimeric human-murine anti-GD2 monoclonal antibody ch14.18 (16), human anti-GD2 monoclonal antibody hu14.18 and human anti-GD2 monoclonal antibody hu3F8 (17). Simon et al (18) identified in a phase IV clinical trial that patients who receive ch14.18 maintenance treatment after chemotherapy acquire a higher 3-year overall survival compared with those who receive only small doses of maintenance chemotherapy or no maintenance therapy.

Recently, numerous studies have focused on enhancing the treatment effects of anti-GD2 monoclonal antibody (1921). The mechanisms by which anti-GD2 monoclonal antibody induces apoptosis of NB include complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) (22). However, numerous studies have suggested that the effect of CDC is associated with side effects, such as pain in anti-GD2 monoclonal antibody treatment (23,24). Sorkin et al (25) identified that a mutation in anti-GD2 monoclonal antibody can reduce pain so that the tolerance to anti-GD2 monoclonal antibody is increased without a reduction in the killing effect of GD2 antibody. Thus, it may be possible to improve the curative effect of anti-GD2 monoclonal antibody through enhancing ADCC.

The ADCC effect of anti-GD2 monoclonal antibody on NB cells is associated with the Fc receptor (FcR) on killer cells, which combines with the Fc fragment of the anti-GD2 monoclonal antibody, activating ADCC and inducing the apoptosis of NB (26). A number of studies have used anti-GD2 monoclonal antibody combined with granulocyte-macrophage colony stimulating factor (GM-CSF) or interleukin-2 (IL-2), and have demonstrated that combination therapy exerts stronger effects compared with using anti-GD2 monoclonal antibody alone (2729). This indicates that an increase in the number or activity of killer cells is a key factor in enhancing the efficacy of anti-GD2 monoclonal antibody.

With the development of tumor immunology in recent years, it has been reported that cytokine-induced killer (CIK)/natural killer (NK) cells transfusion, a kind of adoptive cellular immunotherapy, has significant effect in neuroblastosma MRD treatment without obvious side effects (30). It recognizes and kills target tumor cells by binding specific cell surface markers (31). NK cells are one of the most important immune effector cell types in the process of ADCC (32). However, it has been reported (33) that NK function is limited in some patients with neuroblastoma; therefore, the treatment effect of anti-GD2 monoclonal antibody is reduced in these patients (34). CIK cells are ex vivo-expanded T lymphocytes (35), a subset of T lymphocytes with a natural killer T-cell phenotype expressing both the CD56 and CD3 markers presents non-histocompatibility complex cytotoxicity against target cells, and exhibits improved anti-tumor activity compared with NK or lymphokine-activated killer cells (36).

In our previous study (37), mononuclear cells (MNCs) from cord blood (CBMNCs) were incubated with IL-2 and IL-7 for 21 days, and the percentage of CD3+CD56+ CIK cells and CD3CD56+ NK cells reached 14.26±1.15 and 29.52±0.89%, respectively, while percentage of total CIK/NK (CD3+/−CD56+) cells reached 43.77±1.93%, which increased significantly compare to 9.31±1.77% before culture. Furthermore, the percentage of CD3+CD8+ cells was not significantly increased, indicating a risk reduction of having graft versus host disease (GVHD) in patients. It was also identified that FcRγIII (CD16) and lymphocyte function-associated antigen-1 (LFA-1) are highly expressed in these cells. In conclusion, CIK/NK cells can enhance the killing effect of tumor cells by providing the ligands needed. The effect of CIK/NK cells combined with anti-GD2 monoclonal antibody in targeting human NB cells is not yet known, and further studies are required.

To examine the effects of CIK/NK cells combined with anti-GD2 monoclonal antibody in targeting human NB cells, flow cytometry was performed to detect the expression of GD2 and intercellular adhesion molecule-1 (ICAM-1) on the NB cell line SK-N-SH, and CD16 expression on CIK/NK cells. The aim of this study was to investigate the effect of anti-GD2 monoclonal antibody and CIK/NK cells on SK-N-SH cells. The results provide experimental evidence supporting the use of combined treatment with CIK/NK cells and anti-GD2 monoclonal antibody for MDR-NB, in order to improve the prognosis and overall survival of patients.

Materials and methods

Sample collection and cell line culture

Human cord blood samples were obtained from full-term deliveries between January and March 2016 in the Department of Obstetrics, Sun-Yat Sen Memorial Hospital. This study was approved by the Ethics Committee of Sun Yat-sen Memorial Hospital at Sun Yat-sen University (Guangzhou, China). Patients provided informed written consent. A total of 10 patients aged between 24 and 30 years were recruited. Exclusion criteria included premature delivery and high-risk pregnancies. A total of 30 ml human cord blood was collected from each patent and processed immediately or stored in 4°C for <4 h. MNCs, a group of cells with a one lobed nucleus containing two groups of cells, lymphocytes and monocytes, were isolated using a Ficoll-Paque gradient method (38). A total of 35 ml blood were added slowly into a 50 ml centrifuge tube containing 15 ml Ficoll-Paque Premium (Sigma-Aldrich; Merck KGaA), then centrifuge at 400 × g at room temperature for 20 min. The middle layer cells were collected and washed by phosphate buffer saline (HyClone; GE Healthcare Life Sciences) in triplicate, and cultured in X–VIVO medium (HyClone; GE Healthcare Life Sciences) supplemented with 10% fetal bovine serum (HyClone; GE Healthcare Life Sciences), 1% penicillin-streptomycin (Sigma-Aldrich; Merck KGaA), IL-2 (80 ng/ml; Abcam) and IL-7 (40 ng/ml) at 37°C with 5% CO2. Half of the medium was replaced and cytokines were re-applied every 3 days. The human NB SK-N-SH cell line was purchased from the Cell Bank of Sun-Yat Sen Medical School, and cultured in DMEM (HyClone; GE Healthcare Life Sciences) supplemented with 12% fetal bovine serum (HyClone; GE Healthcare Life Sciences) and 1% penicillin-streptomycin (Sigma-Aldrich; Merck KGaA) at 37°C with 5% CO2.

Characterization of GD2, CD54 and CD16 expression and flow cytometry analysis

Cells were harvested for experiments after 21 days in culture. After cell harvesting, the proportion of CD3+CD56+ (CIK) cells, CD3CD56+ (NK) cells and CD3+/−CD56+ (CIK/NK) cells was detected by flow cytometry. The proportion of CIK cells are a group of immune effector cells characterized by the cell surface marker CD3CD56+ and the proportion of NK cells are a type of lymphocytes, which are distinguished by the cell surface marker CD3-CD56+ (39). Both CIK and NK cells play critical role in immune anti-tumor effect (40). The ratio of CD3+CD56+ CIK, CD3-CD56+ NK or total CD3+/-CD56+ CIK/NK cells accounts for of total mononuclear cells, which was calculated by the following formula: Ratio of CIK cells (%)=number of cells (CD3+CD56+)/number of cells (all MNCs); ratio of NK cells (%)=number of cells (CD3CD56+)/number of cells (all MNCs); ratio of total CIK/NK cells (%)=number of cells (CD3+/−CD56+)/number of cells (all MNCs). The cell surface expression of GD2 and CD54 on SK-N-SH cells and CD16 expression on CIK/NK cells was assessed by staining with 5 µl anti-GD2-phyroerythrin (PE; cat. no. 562100) or anti-CD54-PE-Cy5 (cat. no. 555512) together with anti-45-ECD (cat. no. A07784), or anti-CD16- fluorescein isothiocyanate (cat. no. 555406) antibodies for 20 min at 4°C. Anti-CD45-ECD were purchased from Beckman Coulter, Inc., and all other antibodies were purchased from BD Biosciences. Cells were then washed by phosphate buffer saline (HyClone; GE Healthcare Life Sciences) and analyzed using a BD FACSVerse (BD Biosciences). Data were analyzed using FlowJo software (FlowJo 7.6.1).

Determination of saturated concentration of anti-GD2 on SK-N-SH cells

To determine the effects of different anti-GD2 monoclonal antibody doses on SK-N-SH cells, the cell death rate of NB cells that had been treated with anti-GD2 monoclonal antibody was assessed by flow cytometry. As a preliminary experiment, the optimum treatment concentration of anti-GD2 antibody was tested. Cells (1×105) were seeded in a 24-well plate with C-DMEM medium and cultured for 24 h to allow the cells to adhere to the dish, then 0.5, 1, 2, 5, 10, 20 and 50 µg/ml of anti-GD2 antibody was added to each well. IgG1 was added as a control group. After culture for 1 h, cells were harvested using 0.25% trypsin and washed with phosphate-buffered saline (HyClone; GE Healthcare Life Sciences) in triplicate, then cells were stained with GD2-PE/Cy7 antibody for 20 min in the dark and washed. The expression of GD2 on the cell surface was detected using flow cytometry.

Flow cytometry for cell viability assay

For the determination of optimal effector/target (E/T) ratios of CIK/NK cells over SK-N-SH cells, SK-N-SH cells were treated with 0.25% trypsin to detach them from dishes and washed with PBS in triplicate. Cells were labeled with 3 µM 3,3′-dioctadecyloxacarbocyanine perchlorate (DIO; Beyotime Institute of Biotechnology) at 37°C for 30 min, then 1×105 cells were seeded in a 24-well plate. Different E/T ratios of CIK/NK cells were added and mixed with each well of SK-N-SH cells. The numbers of cells added were 1×105 (E/T=1:1), 2×105 (E/T=2:1), 5×105 (E/T=5:1) and 1×106 (E/T=10:1) (24). The mixed cells were co-cultured in 37°C and 5% CO2 for 4 h. The mixed cells were then harvested and washed with PBS in triplicate, and stained with 5 µl propidium iodide (PI; Sigma-Aldrich; Merck KGaA) for 10 min at 4°C. A flow cytometry assay was performed to detect the percentage of late apoptotic and dead SK-N-SH cells (labeled as DIO+).

For the killing effect experiments, cells were treated with IgG only, CIK/NK cells only, anti-GD2 only or CIK/NK combined with anti-GD2. SK-N-SH cells (1×105) in IgG control group, CIK/NK treated group and CIK/NK + anti-GD2 treated group were labeled with 3 µM DIO for 30 min at 37°C, while cells in the anti-GD2 treated group were not stained with DIO. SK-N-SH cells were mono-cultured or co-cultured with CIK/NK cells (optimal-fold relative cell number) in the absence or presence of saturated concentration of anti-GD2 monoclonal antibody for 4 h at 37°C. Cells were washed with PBS in triplicate and apoptotic cells, defined as late apoptotic and necrotic cells, were labeled with 5 µl PI for 10 min at 4°C before performance of flow cytometry assay. Cell viability was determined using flow cytometry on a BD FACSVerse (BD Biosciences). Data were analyzed by FlowJo software. Each of the experiments were conducted individually, and each of them has a specific control group, therefore the graphs were produced differently according to their own control group.

Cell apoptotic rate in IgG treated group, CIK/NK treated group and CIK/NK+anti-GD2 treated group were calculated using the following equation: Apoptotic rate (%)=DIO+PI+ (%)/DIO+ (%) ×100, where DIO+PI+ cells were the dead SK-N-SH cells that were previously labeled with DIO and then labeled with PI, and DIO+ cells were all the SK-N-SH cells, including living and late apoptotic/dead cells.

Statistical analysis

Data were processed using SPSS 20.0 software (IBM Corp.). A Student's t-test or one-way ANOVA with Tukey's post-hoc test was used to compare groups. P<0.05 was considered to indicate a statistically significant difference. Data are presented as the mean ± standard deviation. All experiments were conducted in triplicate and repeated three times.

Results

GD2 and CD54 (ICAM-1) are highly expressed in human neuroblastoma SK-N-SH cells

To characterize the molecular basis of the effect of anti-GD2 monoclonal antibody or CIK/NK cells on human NB SK-N-SH cells, the cell surface expression of GD2 and CD54 (ICAM-1) was assessed using flow cytometry analyses. The results showed that GD2 and CD54 (ICAM-1) were highly expressed in 93.37±1.2% (Fig. 1A and C) and 91.72±2.15% (Fig. 1B and C) on human neuroblastoma SK-N-SH cells.

Anti-GD2 monoclonal antibody induces cell death at a saturated concentration

The results indicate that following treatment with low dose anti-GD2 antibody, (<5 µg/ml), the detection of GD2 on the cell surface decreased from >95% (in the control group) to ~75% (5 µg/ml treatment group; P<0.001; Fig. 2A). This indicates that anti-GD2 antibody effectively combines with GD2 on the cell surface even at concentrations of <5 µg/ml. However, at doses >5 µg/ml, there was no statistically significant difference in GD2 detection between groups (Fig. 2B), suggesting that increasing the anti-GD2 antibody concentration had no effect on SK-N-SH cells. Therefore, the concentration of 5 µg/ml was selected for subsequent experiments. After 4 h of incubation with 5 µg/ml anti-GD2 antibody 3F8 at 37°C and 5% CO2, cells were observed to be larger and rounded in shape compared with cells prior to treatment (Fig. 2C). The results indicate that anti-GD2 antibody treatment led to 8.43±1.06% cell death compared with 0.92±0.05% in the control group (P<0.001; Fig. 2D). This demonstrates that treatment with anti-GD2 antibody significantly increased apoptosis compared with the control group.

IL-2 and IL-7-cultured CIK/NK cells partially express CD16

CBMNCs were isolated using a density gradient centrifugation method and cultured in complete X–VIVO medium supplemented with IL-2 and IL-7. The proportion of CD3+CD56+ CIK cells and CD3CD56+ NK cells was evaluated prior to culture, and identified to be 2.20±0.92 and 9.85±1.28%, respectively. In total, CD3+/−CD56+ CIK/NK cells accounted for 12.05±2.09%. After 21 days of culture, the proportion of CD3+CD56+ CIK cells increased to 43.60±1.98% and CD3CD56+ NK cells increased to 10.80±1.73%. In total, CD3+/−CD56+ CIK/NK cells accounted for 54.40±1.56% (Fig. 3A and B). These percentages were increased by 19.8-, 1.1- and 4.5-fold compared with before culture, respectively. As shown in Fig. 3C, CBMNCs were single cells when freshly isolated, and cell colonies gradually formed during 21 days of culture. Expression of CD16 on CIK/NK cells was detected after culture, and the results indicated that CD16 was expressed on the cell surface of 19.01±1.27% of CIK/NK cells (Fig. 3D).

CIK/NK cells lead to cell death at various E/T ratios

To determine whether CIK/NK cells could induce cell death in human NB SK-N-SH cell line and to assess the optimal E/T ratio, a specific killing experiment was performed and a flow cytometry assay was used to analyze apoptotic cells. The results showed that after 4 h of culture, CIK/NK cells had gathered around and adhered to targeted NB SK-N-SH cells. The targeted cells had become larger than normal and some of the cell membranes had burst after adherence by CIK/NK cells (Fig. 4A). According to the aforementioned equation, when E/T ratio was 1:1, the proportion of apoptotic cells was 5.94±0.25%. When E/T ratio was increased to 2:1, 5:1 and 10:1, the proportion of apoptotic cells was 8.33±1.46, 8.56±0.71 and 9.00±0.40%, respectively (Fig. 4B and C). One-way ANOVA indicated that when E/T ratio increased from 1:1 to 2:1, the apoptotic rate increased significantly. However, when the E/T ratio was increased further, there were no significant differences between groups (Fig. 4C). This suggested that an E/T ratio of 2:1 was optimal for cell death, and therefore it was selected for subsequent experiments.

Anti-GD2 monoclonal antibody combined with CIK/NK cells significantly increases apoptotic rate

To further investigate whether combining CIK/NK cells with anti-GD2 monoclonal antibody could enhance the induction of cell death, a combination killing experiment was performed. After incubation with an E/T ratio 2:1 of CIK/NK cells and 5 µg/ml anti-GD2 antibody 3F8 at 37°C and 5% CO2 for 4 h, SK-N-SH cells were observed to be surrounded and adhered by CIK/NK cells (Fig. 5A). All cells were subsequently harvested and analyzed by flow cytometry. The results showed that the combined treatment induced an apoptotic rate of 16.92±0.38%, which was significantly higher than the IgG control group and the CIK/NK cell or anti-GD2 antibody mono-treatment groups (P<0.001; Fig. 5B and C).

Discussion

NB is one of the most common pediatric extracranial solid tumors, and its occurrence and development has marked heterogeneity (41). NB can either differentiate from mature nerve cells or low-stage ganglion tumors spontaneously, or metastasize early with highly malignant characteristics (8). Among high-risk stage IV patients with NB, >60% eventually die of relapse even after comprehensive treatment including surgery, radiotherapy or chemotherapy (42). The reason for relapse in patients with stage IV NB is the presence of chemotherapy-resistant NB cells, which induces MRD (5). Chemotherapy is the most effective treatment for NB; however, its effectiveness is constrained by the presence of MDR in tumor cells. Furthermore, increasing the dose of chemotherapeutic drugs may lead to severe toxic effects in multiple organs, including the heart, liver, kidney and ear (43). This suggests that it is necessary to identify a more effective and tolerable treatment strategy for MDR NB.

In recent years, immunotargeting treatment of MDR NB has been demonstrated to significantly prolong patient survival (44). Following treatment with anti-GD2 monoclonal antibody combined with IL-2, GM-CSF or cis-retinoic acid, the 5-year overall survival time of relapsed/refractory patients with NB has improved by 20–30% (45,46). This finding suggests that anti-GD2 monoclonal antibody has enormous potential for the treatment of relapsed/refractory NB. Anti-GD2 monoclonal antibody has been found to stimulate a CDC killing effect by specifically binding to tumor cell surface GD2 or activating the ADCC effect by binding to Fc receptors on killer cells (1723). However, the dosage of anti-GD2 antibody is restricted by severe side effects, such as intolerable pain and allergy (47). This suggests that a novel strategy based on anti-GD2 antibody is required.

CIK cells are mononuclear cells that can be isolated from peripheral blood or cord blood. The cells are cultured with various cytokines and are CD3+CD56+ (48). Due to the expression of CD3 and CD56 on the cell surface, CIK cells not only demonstrate anti-tumor activity, but also have non-major histocompatibility complex-restricted killing activity (49). The killing effect of CIK cells on tumor cells is mainly via expression of LFA-1, which specifically binds to ICAM-1 (CD54) on the surface of tumor cells, resulting in the secretion of benzyloxycarbonyl-L-lysine thiobenzyl ester esterase (granzyme A) that can penetrate the cell membrane and lead to tumor cell death (50). However, previous experiments have demonstrated that the efficacy of CIK/NK cells alone for the treatment of patients with high-risk stage IV NB is limited (51). This is due to the large amount of complement fragments produced by anti-GD2 antibodies during the killing of NB tumor cells, which can cause severe pain (52). Since anti-GD2 is already in late phase of clinical trials (25), numerous evidence has reported that it is effective for the treatment of neuroblastoma, despite of the tolerable side effects, and it shows no harm to normal cells. CIK/NK cell transfusion is a type of adoptive cellular immunotherapy (35), which is commonly used in clinical treatment. It recognizes and kills target tumor cells by binding to specific cell surface markers based on the immune system (17). To the best of our knowledge, there is no evidence showing that CIK/NK cells identify and kill normal cells. Therefore, maximizing the anti-GD2 antibody-mediated ADCC effect is a feasible research direction for improving anti-GD2 antibody treatment for relapsed/refractory NB.

In the present study, the cytokine combination of IL-2 and IL-7 was used for culturing CBMNCs to obtain CIK/NK cells, based on our previous study (37). After 21 days, the proportion of CD3+CD56+CIK and CD3CD56+ NK cells was 43.60±1.98 and 10.80±1.73%, respectively, and the proportion of CIK/NK mixed cells was 54.40±1.56%, which indicated increases of 19.8-, 1.1- and 4.5-fold, respectively. CD16 expression on the surface of CIK/NK cells was 19.01±1.27% after culture, which was similar to the results reported by Bonanno et al (53). Compared with the ‘classical’ CIK amplification system (54) (IL-2, IL-1a, IFN-γ and anti-CD3 monoclonal antibodies), not only were NK cells effectively expanded, but also the proportion of CD8+ cells had not been significantly increased, which could lead to a decreased rate of GVHD (55). This suggests that the two-cytokine culture system provides effective targeting of anti-GD2 antibodies to NB cells and involves a simple preparation process with fewer side effects.

The findings of the present study showed that when various E/T ratios of CIK/NK cells were implemented, the higher the E/T ratio, the more CIK/NK cells adhered around NB tumor cells. After 4 h of treatment, some NB cells exhibited cell membrane rupture and died, indicating that CIK/NK cells had anti-tumor activity against NB cells in vitro. The killing rate gradually increased as E/T ratio was increased, from 5.94±0.26% at the 1:1 ratio to 9.00±0.40% at the 10:1 ratio, suggesting that an increase in effector cells could enhance the anti-tumor effect to a certain extent. It was also found that at E/T >2:1, the cell apoptotic rate curve began to stabilize, suggesting that when CIK/NK cells reached a certain threshold, they could not further increase the apoptotic rate of NB tumor cells. Therefore, clinical application of CIK/NK cell immunotherapy for NB tumors should use an infusion of an appropriate number of cells, in order to achieve maximum efficacy, while minimizing side effects, including GVHD caused by infusion of immune cells.

For the anti-GD2 antibody experiment, it was identified that GD2 was highly expressed on SK-N-SH cells, which was also highly expressed on other malignancies, including bladder cancer (56), breast cancer (57) and some other solid tumors in children (58). The effects of different concentrations of anti-GD2 antibody on NB cells was examined. The experimental results showed that when the antibody concentration reached 5 µg/ml, there were no significant changes in the expression rate of anti-GD2 antibody on the cell surface with increases in concentration. This finding was similar to the results of Esser et al (59). Anti-GD2 antibody mediates the ADCC effect of immune cells on tumor cells in vivo; the Fc segment of anti-GD2 antibody binds to the FcR on the immune cell surface and stimulates the immune cell to release perforin and granzyme that can cleave the NB cell membrane (20). In addition, the anti-GD2 antibody itself can also form antigen-antibody complexes by binding to GD2 antigen on the surface of NB cells, activating the classical complement pathway (CDC pathway) for cell death (60). This study's results showed that the apoptotic rate under anti-GD2 monoclonal antibody alone was 8.43±1.06% at 5 µg/ml. However, activation of the classical complement pathway requires the binding of two or more antibodies to the Fc segment (61), therefore anti-GD2 antibody killing of target cells through the CDC pathway is not a highly effective therapeutic method, which is consistent with our in vitro results.

However, CIK/NK cells combined with anti-GD2 antibody only increased the cell death rate to 16.92±0.38%, which was <20% in the present study. This may be explained by the low expression of FcRγIII (CD16) on the surface of CIK/NK cells, which was only 19.01±1.27%. FcRγIII (CD16) activates the ADCC pathway by combining with Fc region of the anti-GD2 antibody (62). However, this study's bi-factor (IL-2 and IL-7) culture system has no effect on increasing the expression of CD16 on these cells, therefore no comparison is necessary for the expression of CD16 before and after culture. The expression of CD16 on CIK/NK cells was tested after culture, in order to determine the possible proportion of CIK/NK cells involved in anti-GD2-mediated ADCC effect, which may provide an explanation to the low killing effect of combined therapy. Seidel et al (63) showed long treatment time may be beneficial for increasing killing efficacy. Furthermore, in this experiment we defined late apoptosis and cell death induced by CIK/NK cells only, anti-GD2 only or CIK/NK cells combined with anti-GD2 as effective killing, which does not include early apoptosis, therefore PI single staining was performed instead of Annexin-V/PI double staining, which may give another explanation to the low killing effect.

In conclusion, this study provided supporting evidence for treating relapsed/refractory NB with CIK/NK cells combined with anti-GD2 antibodies. Immunotherapy combined with monoclonal antibodies is a potential novel strategy for treating relapsed and refractory NB; however, further studies are required in order to validate this method.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the Guangdong Natural Science Foundation (grant nos. 2014A030313024 and 2017A030313806) and the Guangdong Science and Technology Department (grant no. 2015B050501004).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

CZ and XX performed the experiments and wrote the manuscript. KH designed the experiments and collected the samples. XP analyzed the experimental data. YL, LL and WW performed data analysis and revised the manuscript.

Ethics approval and consent to participate

This study was approved by the Ethics Committee of Sun Yat-sen Memorial Hospital at Sun Yat-sen University (Guangzhou, China). This study was performed in compliance with the Helsinki Declaration. All patients gave written informed consent to participate in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Keshelava N, Davicioni E, Wan Z, Ji L, Sposto R, Triche TJ and Reynolds CP: Histone deacetylase 1 gene expression and sensitization of multidrug-resistant neuroblastoma cell lines to cytotoxic agents by depsipeptide. J Natl Cancer Inst. 99:1107–1119. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Chowdhury F, Lode HN, Cragg MS, Glennie MJ and Gray JC: Development of immunomonitoring of antibody-dependent cellular cytotoxicity against neuroblastoma cells using whole blood. Cancer Immunol Immunother. 63:559–569. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Cotterill SJ, Pearson AD, Pritchard J, Foot AB, Roald B, Kohler JA and Imeson J: Clinical prognostic factors in 1277 patients with neuroblastoma: Results of the European neuroblastoma study group ‘survey’ 1982–1992. Eur J Cancer. 36:901–908. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Maris JM: Recent advances in neuroblastoma. N Engl J Med. 362:2202–2211. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Yamamoto N, Kozaki A, Hartomo TB, Yanai T, Hasegawa D, Kawasaki K, Kosaka Y, Matsuo M, Hirase S, Mori T, et al: Differential expression of minimal residual disease markers in peripheral blood and bone marrow samples from high-risk neuroblastoma patients. Oncol Lett. 10:3228–3232. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Carr-Wilkinson J, O'Toole K, Wood KM, Challen CC, Baker AG, Board JR, Evans L, Cole M, Cheung NK, Boos J, et al: High frequency of p53/mdm2/p14arf pathway abnormalities in relapsed neuroblastoma. Clin Cancer Res. 16:1108–1118. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Gillet JP, Efferth T and Remacle J: Chemotherapy-induced resistance by atp-binding cassette transporter genes. Biochim Biophys Acta. 1775:237–262. 2007.PubMed/NCBI

8 

Jackson JR, Kim Y, Seeger RC and Kim ES: A novel minimal residual disease model of neuroblastoma in mice. J Pediatr Surg. 51:991–994. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Tchirkov A, Greze V, Plantaz D, Rouel N, Vago P and Kanold J: Very long-term molecular follow-up of minimal residual disease in patients with neuroblastoma. Pediatr Blood Cancer. 65:e274042018. View Article : Google Scholar : PubMed/NCBI

10 

Haber M, Smith J, Bordow SB, Flemming C, Cohn SL, London WB, Marshall GM and Norris MD: Association of high-level mrp1 expression with poor clinical outcome in a large prospective study of primary neuroblastoma. J Clin Oncol. 24:1546–1553. 2006. View Article : Google Scholar : PubMed/NCBI

11 

D'Aguanno S, D'Alessandro A, Pieroni L, Roveri A, Zaccarin M, Marzano V, De Canio M, Bernardini S, Federici G and Urbani A: New insights into neuroblastoma cisplatin resistance: A comparative proteomic and meta-mining investigation. J Proteome Res. 10:416–428. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Iwasaki I, Sugiyama H, Kanazawa S and Hemmi H: Establishment of cisplatin-resistant variants of human neuroblastoma cell lines, tgw and goto, and their drug cross-resistance profiles. Cancer Chemother Pharmacol. 49:438–444. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Hakomori S: Tumor-associated carbohydrate antigens defining tumor malignancy: Basis for development of anti-cancer vaccines. Adv Exp Med Biol. 491:369–402. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Allende ML and Proia RL: Lubricating cell signaling pathways with gangliosides. Curr Opin Struct Biol. 12:587–592. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Regina Todeschini A and Hakomori SI: Functional role of glycosphingolipids and gan-gliosides in control of cell adhesion, motility, and growth, through glycosy-naptic microdomains. Biochim Biophys Acta. 1780:421–433. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Wu ZL, Schwartz E, Seeger R and Ladisch S: Expression of GD2 ganglioside by untreated primary human neuroblastomas. Cancer Res. 46:440–443. 1986.PubMed/NCBI

17 

Ahmed M and Cheung NK: Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy. FEBS Lett. 588:288–297. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Simon T, Hero B, Faldum A, Handgretinger R, Schrappe M, Niethammer D and Berthold F: Consolidation treatment with chimeric anti-GD2-antibody ch14.18 in children older than 1 year with metastatic neuroblastoma. J Clin Oncol. 22:3549–3557. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Sait S and Modak S: Anti-GD2 immunotherapy for neuroblastoma. Expert Rev Anticancer Ther. 17:889–904. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Tran HC, Wan Z, Sheard MA, Sun J, Jackson JR, Malvar J, Xu Y, Wang L, Sposto R, Kim ES, et al: TGFβR1 blockade with galunisertib (LY2157299) enhances anti-neuroblastoma activity of the anti-GD2 antibody dinutuximab (ch14.18) with natural killer cells. Clin Cancer Res. 23:804–813. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Ladenstein R, Pötschger U, Valteau-Couanet D, Luksch R, Castel V, Yaniv I, Laureys G, Brock P, Michon JM, Owens C, et al: Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol. 19:1617–1629. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Modak S and Cheung NK: Disialoganglioside directed immunotherapy of neuroblastoma. Cancer Invest. 25:67–77. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Cheung NK, Cheung IY, Kushner BH, Ostrovnaya I, Chamberlain E, Kramer K and Modak S: Murine anti-GD2 monoclonal antibody 3F8 combined with granulocyte-macrophage colony-stimulating factor and 13-cis-retinoic acid in high-risk patients with stage 4 neuroblastoma in first remission. J Clin Oncol. 30:3264–3270. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Federico SM, McCarville MB, Shulkin BL, Sondel PM, Hank JA, Hutson P, Meagher M, Shafer A, Ng CY, Leung W, et al: A pilot trial of humanized anti-GD2 monoclonal antibody (hu14.18K322A) with chemotherapy and natural killer cells in children with recurrent/refractory neuroblastoma. Clin Cancer Res. 23:6441–6449. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Sorkin LS, Otto M, Baldwin WM III, Vail E, Gillies SD, Handgretinger R, Barfield RC, Ming Yu H and Yu AL: Anti-GD(2) with an FC point mutation reduces complement fixation and decreases antibody-induced allodynia. Pain. 149:135–142. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Xu H, Guo H, Cheung IY and Cheung NK: Antitumor efficacy of anti-GD2 IgG1 is enhanced by fc glyco-engineering. Cancer Immunol Res. 4:631–638. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Cheung IY, Kushner BH, Modak S, Basu EM, Roberts SS and Cheung NV: Phase I trial of anti-GD2 monoclonal antibody hu3F8 plus GM-CSF: Impact of body weight, immunogenicity and anti-GD2 response on pharmacokinetics and survival. Oncoimmunology. 6:e13583312017. View Article : Google Scholar : PubMed/NCBI

28 

Kushner BH, Cheung IY, Modak S, Basu EM, Roberts SS and Cheung NK: Humanized 3F8 anti-GD2 monoclonal antibody dosing with granulocyte-macrophage colony-stimulating factor in patients with resistant neuroblastoma: A phase 1 clinical trial. JAMA Oncol. 4:1729–1735. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Ozkaynak MF, Gilman AL, London WB, Naranjo A, Diccianni MB, Tenney SC, Smith M, Messer KS, Seeger R, Reynolds CP, et al: Corrigendum: A comprehensive safety trial of chimeric antibody 14.18 with GM-CSF, IL-2, and isotretinoin in high-risk neuroblastoma patients following myeloablative therapy: Children's oncology group study ANBL0931. Front Immunol. 9:16412018. View Article : Google Scholar : PubMed/NCBI

30 

Shoae-Hassani A, Hamidieh AA, Behfar M, Mohseni R, Mortazavi-Tabatabaei SA and Asgharzadeh S: NK cell-derived exosomes from NK cells previously exposed to neuroblastoma cells augment the antitumor activity of cytokine-activated NK cells. J Immunother. 40:265–276. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Zenarruzabeitia O, Vitallé J, Astigarraga I and Borrego F: Natural killer cells to the attack: Combination therapy against neuroblastoma. Clin Cancer Res. 23:615–617. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Muntasell A, Ochoa MC, Cordeiro L, Berraondo P, López-Díaz de Cerio A, Cabo M, López-Botet M and Melero I: Targeting NK-cell checkpoints for cancer immunotherapy. Curr Opin Immunol. 45:73–81. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Burga RA, Yvon E, Chorvinsky E, Fernandes R, Cruz CRY and Bollard CM: Engineering the TGFβ receptor to enhance the therapeutic potential of natural killer cells as an immunotherapy for neuroblastoma. Clin Cancer Res. 25:4400–4412. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Thommesen JE, Michaelsen TE, Loset GA, Sandlie I and Brekke OH: Lysine 322 in the human IgG3 C (H)2 domain is crucial for antibody dependent complement activation. Mol Immunol. 37:995–1004. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Schmeel LC, Schmeel FC, Coch C and Schmidt-Wolf IG: Cytokine-induced killer (CIK) cells in cancer immunotherapy: Report of the international registry on CIK cells (IRCC). J Cancer Res Clin Oncol. 141:839–849. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG and Weissman IL: Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med. 174:139–149. 1991. View Article : Google Scholar : PubMed/NCBI

37 

Wu YF, Lin YC, LI Y, Wang XP and Wei J: Exploratory research of setting an optimal minor-factor culture system of cytokine-induced killer cells/natural cells. J Sun Yat-Sen University (Medical Sciences),. 30:361–366. 2009.

38 

Akhter N, Madhoun A, Arefanian H, Wilson A, Kochumon S, Thomas R, Shenouda S, Al-Mulla F, Ahmad R and Sindhu S: Oxidative stress induces expression of the toll-like receptors (TLRs) 2 and 4 in the human peripheral blood mononuclear cells: Implications for metabolic inflammation. Cell Physiol Biochem. 53:1–18. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Biederbick KD and Schmidt-Wolf IGH: Efficacy of cytokine-induced killer cells targeting CD40 and GITR. Oncol Lett. 17:2425–2430. 2019.PubMed/NCBI

40 

Gao D, Cai Y, Chen Y, Li W, Wei CC, Luo X and Wang Y: Novel TLR7 agonist stimulates activity of CIK/NK immunological effector cells to enhance antitumor cytotoxicity. Oncol Lett. 15:5105–5110. 2018.PubMed/NCBI

41 

Pinto NR, Applebaum MA, Volchenboum SL, Matthay KK, London WB, Ambros PF, Nakagawara A, Berthold F, Schleiermacher G, Park JR, et al: Advances in risk classification and treatment strategies for neuroblastoma. J Clin Oncol. 33:3008–3017. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Weinstein JL, Katzenstein HM and Cohn SL: Advances in the diagnosis and treatment of neuroblastoma. Oncologist. 8:278–292. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Ning BT, Yu B, Chan S, Chan JL, Huang JD and Chan GC: Treatment of neuroblastoma with an engineered ‘Obligate’ anaerobic salmonella typhimurium strain YB1. J Cancer. 8:1609–1618. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Kholodenko IV, Kalinovsky DV, Doronin II, Deyev SM and Kholodenko RV: Neuroblastoma origin and therapeutic targets for immunotherapy. J Immunol Res. 2018:73942682018. View Article : Google Scholar : PubMed/NCBI

45 

Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, et al: Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 363:1324–1334. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Dobrenkov K and Cheung NK: GD2-targeted immunotherapy and radioimmunotherapy. Semin Oncol. 41:589–612. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Anghelescu DL, Goldberg JL, Faughnan LG, Wu J, Mao S, Furman WL, Santana VM and Navid F: Comparison of pain outcomes between two anti-GD2 antibodies in patients with neuroblastoma. Pediatr Blood Cancer. 62:224–228. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Pan Y, Wu Y, Ji J, Cai H, Wang H, Jiang Y, Sang L, Yang J, Gao Y, Liu Y, et al: Effect of cytokine-induced killer cells on immune function in patients with lung cancer. Oncol Lett. 11:2827–2834. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Sangiolo D, Martinuzzi E, Todorovic M, Vitaggio K, Vallario A, Jordaney N, Carnevale-Schianca F, Capaldi A, Geuna M, Casorzo L, et al: Alloreactivity and anti-tumor activity segregate within two distinct subsets of cytokine-induced killer (CIK) cells: Implications for their infusion across major HLA barriers. Int Immunol. 20:841–848. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Kim HM, Lim J, Kang JS, Park SK, Lee K, Kim JY, Kim YJ, Hong JT, Kim Y and Han SB: Inhibition of human cervical carcinoma growth by cytokine-induced killer cells in nude mouse xenograft model. Int Immunopharmacol. 9:375–380. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Xu YC, Xu Q, Li JJ, Gu XF, Lin XL, Sun L, Lu HM, Tang L, Ma Y, Lu Z and Wang HX: Chemotherapy with or without autologous cytokine-induced killer cell transfusion as the frst-line treatment for stage IV gastrointestinal cancer: A phase II clinical trial. J Cancer Res Clin Oncol. 142:1315–1323. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Mueller I, Ehlert K, Endres S, Pill L, Siebert N, Kietz S, Brock P, Garaventa A, Valteau-Couanet D, Janzek E, et al: Tolerability, response and outcome of high-risk neuroblastoma patients treated with long-term infusion of anti-GD2 antibody ch14.18/CHO. MAbs. 10:55–61. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Bonanno G, Iudicone P, Mariotti A, et al: Thymoglobulin, interferon-g and interleukin-2 efficiently expand cytokine-induced killer (CIK) cells in clinical-grade cultures. J Transl Med. 8:1292010. View Article : Google Scholar : PubMed/NCBI

54 

Leemhuis T, Wells S, Scheffold C, Edinger M and Negrin RS: A phase I trial of autologous cytokine-induced killer cells for the treatment of relapsed Hodgkin disease and non-Hodgkin lymphoma. Biol Blood Marrow Transplant. 11:181–187. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Rettinger E, Kreyenberg H, Merker M, Kuçi S, Willasch A, Bug G, Ullrich E, Wels WS, Bonig H, Klingebiel T and Bader P: Immunomagnetic selection or irradiation eliminates alloreactive cells but also reduces anti-tumor potential of cytokine-induced killer cells: Implications for unmanipulated cytokine-induced killer cell infusion. Cytotherapy. 16:835–844. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Vantaku V, Donepudi SR, Ambati CR, Jin F, Putluri V, Nguyen K, Rajapakshe K, Coarfa C, Battula VL, Lotan Y and Putluri N: Expression of ganglioside GD2, reprogram the lipid metabolism and EMT phenotype in bladder cancer. Oncotarget. 8:95620–95631. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Orsi G, Barbolini M, Ficarra G, Tazzioli G, Manni P, Petrachi T, Mastrolia I, Orvieto E, Spano C, Prapa M, et al: GD2 expression in breast cancer. Oncotarget. 8:31592–31600. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Tesfaye M and Savoldo B: Adoptive cell therapy in treating pediatric solid tumors. Curr Oncol Rep. 20:732018. View Article : Google Scholar : PubMed/NCBI

59 

Esser R, Müller T, Stefes D, Kloess S, Seidel D, Gillies SD, Aperlo-Iffland C, Huston JS, Uherek C, Schönfeld K, et al: NK cells engineered to express a GD2-specific antigen receptor display built-in ADCC-like activity against tumor cells of neuroectodermal origin. J Cell Mol Med. 16:569–581. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Wold ED, Smider VV and Felding BH: Antibody therapeutics in oncology. Immunotherapy (Los Angel). 2:1082016. View Article : Google Scholar : PubMed/NCBI

61 

Stasiłojć G, Österborg A, Blom AM and Okrój M: New perspectives on complement mediated immunotherapy. Cancer Treat Rev. 45:68–75. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Kudo K, Imai C, Lorenzini P, Kamiya T, Kono K, Davidoff AM, Chng WJ and Campana D: T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 74:93–103. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Seidel D, Shibina A, Siebert N, Wels WS, Reynolds CP, Huebener N and Lode HN: Disialoganglioside-specific human natural killer cells are effective against drug-resistant neuroblastoma. Cancer Immunol Immunother. 64:621–634. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang C, Xiong X, Li Y, Huang K, Liu L, Peng X and Weng W: Cytokine‑induced killer cells/natural killer cells combined with anti‑GD2 monoclonal antibody increase cell death rate in neuroblastoma SK‑N‑SH cells. Oncol Lett 18: 6525-6535, 2019
APA
Zhang, C., Xiong, X., Li, Y., Huang, K., Liu, L., Peng, X., & Weng, W. (2019). Cytokine‑induced killer cells/natural killer cells combined with anti‑GD2 monoclonal antibody increase cell death rate in neuroblastoma SK‑N‑SH cells. Oncology Letters, 18, 6525-6535. https://doi.org/10.3892/ol.2019.11020
MLA
Zhang, C., Xiong, X., Li, Y., Huang, K., Liu, L., Peng, X., Weng, W."Cytokine‑induced killer cells/natural killer cells combined with anti‑GD2 monoclonal antibody increase cell death rate in neuroblastoma SK‑N‑SH cells". Oncology Letters 18.6 (2019): 6525-6535.
Chicago
Zhang, C., Xiong, X., Li, Y., Huang, K., Liu, L., Peng, X., Weng, W."Cytokine‑induced killer cells/natural killer cells combined with anti‑GD2 monoclonal antibody increase cell death rate in neuroblastoma SK‑N‑SH cells". Oncology Letters 18, no. 6 (2019): 6525-6535. https://doi.org/10.3892/ol.2019.11020