Enhanced antitumor effects and improved immune status of dendritic cell and cytokine‑induced killer cell infusion in advanced cancer patients

  • Authors:
    • Feng Chen
    • Menghan Yang
    • Qingkun Song
    • Jiangping Wu
    • Xiaoli Wang
    • Xinna Zhou
    • Yanhua Yuan
    • Yuguang Song
    • Ni Jiang
    • Yanjie Zhao
    • Lei Zhou
    • Jun Ren
  • View Affiliations

  • Published online on: September 19, 2017     https://doi.org/10.3892/mco.2017.1415
  • Pages: 903-910
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Little progress has been made in the treatment of advanced cancer. Dendritic cells (DCs) plus cytokine‑induced killer (CIK) cells have exhibited antitumor effects. Thus, the aim of the present study was to evaluate the clinical efficacy of DC‑CIK cell treatment in patients with advanced cancer. A paired study including 57 patients treated with DC‑CIK cells (DC‑CIK group) and 33 patients treated with best supportive care alone (BSC group) was performed. The patients in the DC‑CIK group were matched to those in the control group in terms of sex, age, tumor type and clinical stage. T‑cell subsets were detected and overall survival (OS) was compared between the two groups. The results demonstrated that CD4+/CD25+ and CD8+/CD28‑ subsets significantly decreased following DC‑CIK immunotherapy (P<0.05). The CD3+, CD3+/CD8+, CD8+/CD28+ and CD3+/CD56+ T‑cell subsets were significantly increased in the DC‑CIK group compared with the BSC group, while the CD8+/CD28‑ subset was significantly decreased. Univariate analysis demonstrated that a lower CD8+/CD28‑ and a higher CD8+/CD28+ ratio were associated with prolonged OS in advanced cancer patients. In addition, DC‑CIK treatment administration, age (>60 vs. <60 years), clinical stage and the frequency of CIK treatment significantly affected the OS of patients in the DC‑CIK group. A CD8+/CD28‑ ratio of <21.12 was found to decrease the hazard ratio (HR) of OS to 0.50 [95% confidence interval (CI): 0.29‑0.87] and a CD8+/CD28+ ratio >9.04 was found to decrease the HR of OS to 0.45 (95% CI: 0.21‑0.98). No serious side effects were observed in the DC‑CIK group. Taken together, these data indicate that DC‑CIK infusions were able to change the ratios of the T‑cell subsets, which increased the T helper cell and cytotoxic T lymphocyte subsets, while it decreased regulatory T lymphocyte subsets. Thus, this method of immunotherapy was found to improve the imbalance in the immune system and prolong the OS in patients with advanced cancer.

Introduction

Cancer is among the leading causes of morbidity and mortality worldwide, with an estimated 14 million new cases and 8.2 million cancer-related deaths annually. Over 60% of the cases globally occur in developing countries, which account for ~70% of cancer-related mortality worldwide (1). Given their poor overall condition, patients with late-stage cancer are mostly not eligible for conventional anticancer treatments such as surgery, chemotherapy, or radiotherapy, and best supportive care (BSC) is currently considered as the only option for patients with a relatively poor prognosis. Several efforts have been made to improve the survival of patients with advanced cancer (2). However, the results thus far have been unsatisfactory. Therefore, further efforts must be made to improve the current therapeutic modalities and to explore novel therapies for advanced cancer, in order to improve patient care and prolong survival.

Immunotherapy has been shown to be effective in the treatment of a number of malignant tumors, with adoptive cellular immunotherapy being considered a promising and effective modality (38). Several types of immune cells, such as lymphokine-activated killer cells (9), tumor-infiltrating lymphocytes (10) and anti-CD3 monoclonal antibody-induced killer cells (11) have shown efficacy in advanced cancers. Among these cells, cytokine-induced killer (CIK) cells have several advantages compared with traditional immune cells, such as proliferating rapidly in vitro, exhibiting intensified antitumor activity and a broader spectrum of targeted tumors, and being associated with less severe side effects, which qualifies them as one of the most promising treatments, particularly for patients with advanced cancer (6,1217). It is noteworthy that the antitumor activity of CIK cells may be activated and enhanced by dendritic cells (DCs), which are the main antigen-presenting cells (18,19). DCs present tumor antigens through MHCII molecules, preventing the immune escape of tumor cells. CIK cells are also able to recognize DCs in a T-cell receptor-independent manner, and the DC-CIK interaction stimulates the proliferation and antitumor activity of CIK cells through secreting interleukin (IL)-12, interferon (IFN)-γ, and other cytokines (20). DCs plus CIK cells not only enhance the antitumor effect, but also regulate and improve the immune function in cancer patients (2022). Lately, a report from the International Registry on CIK cells found that adjuvant immunotherapy with CIK cells may prevent recurrence and improve the quality of life and progression-free survival rates in cancer patients (6). Our previous study also indicated that DC-CIK treatment may be able to recover cellular immunity and improve the Eastern Cooperative Oncology Group (ECOG) performance status and quality of life in patients with advanced cancer (23,24).

Current data from clinical studies on the antitumor effects and prognostic benefits of DC-CIK cells are limited, particularly for patients with advanced cancer. The aim of the present study was to evaluate the clinical efficacy of DC-CIK cell treatment in patients with advanced cancer.

Patients and methods

Patients

A paired study was performed to compare the clinical outcomes of advanced cancer patients received either autologous DC-CIK immunotherapy or BSC alone. Between June 2012 and January 2014, a total of 90 patients with advanced cancer were recruited in the present study from the Beijing Shijitan Hospital Cancer Center (Capital Medical University, Beijing, China). A total of 57 patients underwent DC-CIK immunotherapy (DC-CIK group) and 33 patients were administered BSC alone (BSC group). All the patients had a definitive histological or cytological diagnosis and were unresponsive or intolerant to conventional anticancer treatments, such as surgery, chemotherapy or radiotherapy. The characteristics of the patients are summarized in Table I. The criteria for patient selection were as follows: i) Patient aged 18–85 years; ii) chemoradiotherapy-free for ≥3 months; iii) expected survival duration of >3 months; iv) ECOG performance status of 0–2; v) white blood cell (WBC) count >3,500/µl; vi) hemoglobin level >80 g/dl; vii) platelet count >100,000/µl; viii) serum aspartate aminotransferase (AST)/alanine aminotransferase (ALT) <2.0 the upper limit of normal; ix) no cardiac arrhythmias, congestive heart failure or severe coronary artery disease; x) no active autoimmune disease or T-cell lymphoma; and xi) no pregnancy or lactation. The subjects in the two groups were matched for sex, age, stage, pathology, tumor size and metastasis at the first visit.

Table I.

Patient characteristics.

Table I.

Patient characteristics.

CharacteristicsDC-CIK groupBSC groupP-value
No. of patients5733
Sex (n)0.230
  Male2720
  Female3013
Age (years)0.874
  Mean ± standard deviation60.00±16.3060.52±11.64
  <60 (n)3116
  ≥60 (n)2617
Tumor types (n)0.450
  Digestive system cancer1911
  Lung cancer and mesothelioma1211
  Breast cancer83
  Head and neck cancer42
  Male genitourinary system cancer41
  Female genitourinary system cancer30
  Lymphoma35
  Sarcoma20
  Glioblastoma10
  Melanoma10
Stage (n) 0.105
  IV4019
  III1511
  II23
DC-CIK treatment cycles (n)
  133
  211
  37
  ≥46

[i] DC-CIK, dendritic cell/cytokine-induced killer cell immunotherapy; BSC, best supportive care.

Informed consent was obtained from all the patients. After enrollment, a complete medical history was taken and physical examination was conducted by professional oncologists for each patient. This study was approved by the Ethics Committee of the Beijing Shijitan Hospital.

Treatment

Patients in the control group received BSC alone, which included active symptom control, pain management, and the multiprofessional attention to the individual's overall physical, psychosocial, spiritual and cultural needs. Patients in the DC-CIK group received autologous DC-CIK cells with an interval of 1 month in addition to BSC. For each cycle of treatment, the patients received three intravenous infusions of DC-CIK cells with 1-day intervals. Patients without disease progression were eligible for maintenance treatment. For each cycle of treatment, the patients received a median of 6.47×108 (range, 5.35×107-2.98×109) of autologous DC cells and 7.35×109 (range, 3.00–17.25×109) of CIK cells. The median number of CIK cell immunotherapy cycles was 2 (range, 1–13 cycles). All the patients were seen biweekly or monthly by oncology specialists, and clinical examinations were performed monthly, including physical examination, T-cell subsets, routine blood count, serum AST and ALT, blood urea nitrogen and creatinine, and electrocardiogram. Additional care was provided if needed.

Clinical assessment of response and toxicity

All the patients were followed up at the outpatient clinic or the oncology ward from the date of initial treatment to March 31, 2016, or to the date of death. Clinical response was determined according to the National Cancer Institute's Response Evaluation Criteria in Solid Tumors, version 1.1 (https://ctep.cancer.gov/protocoldevelopment/docs/recist_guideline.pdf). Patients were assessed by oncologists after each cycle of treatment, including color Doppler ultrasound, computed tomography scan, magnetic resonance imaging and technetium bone scan. Overall survival (OS) was calculated from the time of treatment initiation until death, and patients who remained alive were censored at the time of the last follow-up. Adverse events were evaluated according to the World Health Organization (WHO) criteria (25).

Preparation of DC and CIK cells

CIK cells were prepared as previously described (23). Briefly, peripheral blood mononuclear cells (PBMCs) were mobilized by granulocyte colony-stimulating factor (G-CSF) until WBC ≥10,000/µl, lymphocytes + monocytes ≥15%. Apheresis was performed from the patients using the COBE Spectra cell separator (COBE BCT, Lakewood, CO, USA) and repeated until ≥4.5×106/kg CD34+ cells were collected. PBMCs were separated by the Ficoll-Hypaque centrifugation method and incubated for 2 h, and the adherent cells were cultured in vitro to generate autologous DCs in the presence of IL-4, tumor necrosis factor-α and granulocyte-macrophage (GM)-CSF (Boehringer, Mannheim, Germany). For the culture of autologous CIKs, PBMCs were cultured in AIM-V medium containing the recombinant cytokines IL-2, IFN-γ and monoclonal anti-human CD3 antibody (50 ng/ml; IM1650, Boehringer). The cells were incubated in a humidified atmosphere with 5% CO2 at 37°C. IL-2 and IFN-γ were added to the medium every 5 days.

Cell growth was observed under the microscope, and DC phenotypes were determined by flow cytometry of CD80, CD86, HLA-DR, CD1a and CD11c (Beckman-Coulter, Shanghai, China). The DC suspension contained >80% of CD80+/CD86+ cells prior to infusion. The CIKs expressed CD3 and CD56 (Beckman-Coulter). After culture in vitro for 7–10 days, DCs and CIKs were harvested and administered intravenously 3 times with 1-day intervals.

Detecting the phenotype of DCs, CIK cells and T-cell subsets

The phenotype of DCs and CIK cells was determined prior to infusion. T-cell subsets were assessed at the beginning of the first treatment and redetected monthly. Briefly, 2 ml of heparinized peripheral blood was drawn from each patient and PBMCs were separated by the Ficoll-Hypaque centrifugation method. A total of 100 µl PBMCs were incubated in the dark with primary antibody at 4°C for 15 min. After hemolysis for 10 min, the samples were centrifuged for 10 min at 450 × g at room temperature, and then washed twice in phosphate-buffered saline and subjected to flow cytometric analysis (Becton-Dickinson, Franklin Lakes, NJ). The primary antibodies included: CD4-FITC (A07550), CD8-PE (IM1650), CD3-PerCP (A07749), CD25-PE (A07774), CD28-FITC (IM1236U), CD3-FITC (A07746) and CD56-PE (IM2073U) (Beckman-Coulter). All the antibodies were mouse anti-human monoclonal antibodies, with 1:10 dilution.

The cell phenotypes were analyzed by flow cytometry (FC500, Beckman-Coulter) and CXP analysis software (Beckman-Coulter) was used. Lymphocyte subset levels were reported as percentages of the total population.

Statistical analysis

Data were analyzed using the SPSS 16.0 software package (SPSS Inc., Chicago, IL, USA). T-cell subsets were expressed as mean ± standard deviation. The independent samples t-test and paired t-test were used to compare the changes in T-cell populations between the two groups. The OS rate and survival curves were calculated by the Kaplan-Meier method. Associations between OS and potential prognostic factors were estimated using the log-rank test in univariate analyses. The significant variables were further analyzed by the Cox hazard proportional regression model with adjustments for age, sex and tumor type. All tests were two-sided and the significance level was set at 0.05.

Results

Patient characteristics

Of the 57 patients in the DC-CIK group, 27 were male and 30 were female; the mean patient age was 60 years (range, 6–81 years). The patient characteristics are summarized in Table I. For each cycle of treatment, the patients received three intravenous infusions of DC-CIK cells with 1-day intervals. The median number of CIK cell immunotherapy cycles was 2 (range, 1–13 cycles).

Of the 33 patients in the BSC group, 20 were male and 13 were female; the mean patient age was 61 years (range, 42–85 years). The patients were administered BSC at the Department of Oncology of our hospital between June 2012 and January 2014.

The patient characteristics, such as sex, age, tumor type and clinical stage, were comparable between the two groups (Table I).

Comparison of T-cell subsets before and after DC-CIK infusion

To investigate the immunomodulatory effects of DC-CIK cell treatment, the T-cell subsets in the 57 patients in the DC-CIK group were analyzed prior to and 1 month after DC-CIK cell infusion. The CD3+, CD3+/CD4+, CD3+/CD8+, CD8+/CD28+ and CD3+/CD56+ T-cell subsets were significantly increased following DC-CIK treatment (P<0.05). Conversely, the CD4+/CD25+, CD8+/CD28 subsets were significantly decreased following DC-CIK immunotherapy (P<0.05) (Table II).

Table II.

T-cell subsets in the peripheral blood before and after DC-CIK cell treatment.

Table II.

T-cell subsets in the peripheral blood before and after DC-CIK cell treatment.

T-cell subsetsBefore treatment (%)After treatment (%)P-value
CD3+ 68.67±11.23 82.55±12.590.000
CD3+/CD4+ 34.23±11.97 40.22±17.090.005
CD3+/CD8+ 32.27±12.21 42.42±17.830.000
CD4+/CD25+ 4.48±3.05 3.23±2.650.024
CD8+/CD28 23.39±10.09 17.97±9.180.000
CD8+/CD28+ 14.11±8.46 27.00±15.100.000
CD3+/CD56+ 10.88±7.76 15.56±16.240.047

[i] Values are presented as mean ± standard deviation. DC-CIK, dendritic cell-cytokine-induced killer cell immunotherapy.

Comparison of the changes in T-cell subsets between the DC-CIK and BSC groups

Next, the changes in the T-cell subsets in peripheral blood between the DC-CIK and BSC groups we observed. The T-cell subsets were analyzed prior to and 1 month after the first treatment in patients from the two groups. Although no significant difference in the T-cell subsets were observed between the two groups at the beginning of the first treatment (data not shown), the CD3+, CD3+/CD8+, CD8+/CD28+ and CD3+/CD56+ T-cell subsets were significantly increased in the DC-CIK group compared with the BSC group, while the CD8+/CD28 subset decreased significantly. No significant differences in the CD3+/CD4+ and CD4+/CD25+ subsets were observed between the two groups before and after treatment (Table III). Thus, these data indicated that DC-CIK cell treatment improved cellular immune function in advanced cancer patients.

Table III.

Comparison of the changes in T-cell subsets between the BSC and DC-CIK groups.

Table III.

Comparison of the changes in T-cell subsets between the BSC and DC-CIK groups.

T cell subsetsBSC group (%)aDC-CIK group (%)aP-value
CD3+ 2.09±10.89 13.88±13.720.000
CD3+/CD4+ 0.85±10.54 5.99±15.550.367
CD3+/CD8+ 0.97±7.38 10.15±18.000.006
CD4+/CD25+ −0.79±4.55 −1.25±4.070.247
CD8+/CD28 1.96±8.68 −5.42±7.850.011
CD8+/CD28+ −0.22±5.20 12.89±14.760.000
CD3+/CD56+ −1.88±5.17 4.67±17.37 0.018

{ label (or @symbol) needed for fn[@id='tfn3-mco-0-0-1415'] } Values are presented as mean ± standard deviation.

a Difference in T-cell subsets before and after treatment. BSC, best supportive care; DC-CIK, dendritic cell-cytokine-induced killer cell immunotherapy.

Association between cycles of DC-CIK infusion and T-cell subset changes

The effect of the frequency of DC-CIK infusion on the changes in T-cell subsets was further evaluated, and it was observed that the T-cell subsets changed after 1 cycle of DC-CIK immunotherapy. CD3+, CD3+/CD4+, CD3+/CD8+, CD8+/CD28+ and CD3+/CD56+ subsets were significantly increased, while CD4+/CD25+ and CD8+/CD28 were significantly decreased. However, no obvious changes in T-cell subsets were observed before or after >2 cycles of infusion (Table IV).

Table IV.

Associations between cycles of DC-CIK infusion and T-cell subset changes.

Table IV.

Associations between cycles of DC-CIK infusion and T-cell subset changes.

Before treatment1 cycle2 cycles3 cycles≥4 cycles
Cases, n 5726136
CD3+ 68.67±11.23 82.55±12.59b 80.87±15.01a 83.59±13.30 86.20±12.54a
CD3+/CD4+ 34.23±11.97 40.22±17.09b 41.17±18.71 29.73±15.75 41.25±8.38a
CD3+/CD8+ 32.27±12.21 42.42±17.83b 29.49±14.27 44.79±9.32 35.88±15.38
CD4+/CD25+ 4.48±3.05 3.23±2.65a 2.35±1.49a 2.61±1.35 3.07±2.42
CD8+/CD28 23.39±10.09 17.97±9.18b 16.98±6.92 22.07±10.95a 15.72±6.58a
CD8+/CD28+ 14.11±8.46 27.00±15.10b 25.94±17.11 18.47±11.32 32.05±12.77a
CD3+/CD56+ 10.88±7.76 15.56±16.24a 24.25±21.96 9.50±5.55 5.73±2.42

a Difference in T-cell subsets before and after treatment (P<0.05).

b Difference in T-cell subsets before and after treatment (P<0.01). Values are presented as mean ± standard deviation. DC-CIK, dendritic cell-cytokine-induced killer cell immunotherapy.

Association between OS and T-cell subset changes

To investigate the factors that affect the OS of patients with advanced cancer, a univariate analysis was conducted, demonstrating that a lower CD8+/CD28 and a higher of CD8+/CD28+ ratio may be associated with longer OS. In addition, DC-CIK treatment administration, age (>60 vs. <60 years), clinical stage and the frequency of CIK treatment significantly affected the OS of patients in the DC-CIK group (Table V, Figs. 1 and 2).

Table V.

Univariate analysis of the patient's clinical characteristics and overall survival.

Table V.

Univariate analysis of the patient's clinical characteristics and overall survival.

VariablesMedian OS (months)Log-rank Relative risk (95% CI)P-value
CD8+/CD28 0.006
  ≥21.1211.008.96–13.04
  <21.1214.0010.88–17.12
CD8+/CD28+ 0.007
  ≤9.0410.007.08–12.92
  >9.0413.0010.82–15.18
DC-CIK therapy 0.021
  Yes11.009.17–12.83
  No14.0011.54–16.46
Age (years) 0.050
  <6015.0010.54–19.46
  ≥6012.0010.51–13.49
Clinical stage 0.032
  II12.00–28.00
  III19.0013.34–20.54
  IV12.0011.05–15.44
DC-CIK cycles (n) 0.040
  011.009.17–12.83
  114.0011.35–16.65
  ≥214.0010.56–17.45

[i] OS, overall survival; CI, confidence interval; DC-CIK, dendritic cell-cytokine-induced killer cell immunotherapy.

The parameters that affected the prognosis of advanced cancer patients in this study were further analyzed through the Cox hazard proportional regression model with adjustments for age, sex and tumor type. A multivariate analysis demonstrated that a CD8+/CD28 ratio <21.12 decreased the hazard ratio (HR) of OS to 0.50 [95% confidence interval (CI): 0.29–0.87] and a CD8+/CD28+ ratio >9.04 decreased the HR of OS to 0.45 (95% CI: 0.21–0.98) (Table VI, Figs. 3 and 4). Thus, taken together, these results demonstrated that T-cell populations may be associated with the OS of patients with advanced cancer, and DC-CIK immunotherapy may improve OS.

Table VI.

Multivariable analysis of the patients' clinical characteristics and overall survival.

Table VI.

Multivariable analysis of the patients' clinical characteristics and overall survival.

ParametersHazard ratio95% CI
CD8+/CD28 <21.120.5000.288–0.866
CD8+/CD28+ >9.040.4350.209–0.907

[i] CI, confidence interval.

Side effects

No severe side effects were observed during DC-CIK cell treatment. Three patients in the DC-CIK group developed fever (<38.5°C) that was spontaneously relieved 2 h after the infusion. No other serious adverse events, such as high fever, chills, rash or hemolytic anemia, were reported in patients receiving DC-CIK cell treatment.

Discussion

Accumulating evidence supports cellular immunotherapy, which directly or indirectly regulates the biological interaction between the host and the tumor (26), as a potential strategy for the improvement of cancer treatment, with CIK cells representing a promising cellular immunotherapy associated with several advantages, such as MHC-unrestricted cytotoxic activity, increase in cytokine secretion, improvement of immune function and induction of apoptosis of cancer cells (27), and may thus be beneficial to patients with advanced cancer.

An increasing amount of studies demonstrated that CIK cell-based immunotherapy is a promising new treatment modality with the potential of improving the prognosis of cancer patients (1824,26,27). A study from the International Registry reported the results of 11 CIK cell treatment trials for a variety of cancers, including hepatocellular carcinoma, gastric cancer and Hodgkin or non-Hodgkin lymphoma, and demonstrated a total response rate of 91/384 reported patients; 161 patients had stable disease and 129 patients had progressive disease. The disease-free survival rates were significantly higher in patients treated with CIK cells compared with those in the control group without CIK treatment (6). The present study demonstrated that the OS favored the DC-CIK arm compared with the BSC arm. The results indicated a 3-month prolongation in the median OS in favor of the DC-CIK treatment compared with the BSC alone arm (14.00 vs. 11.00 months, respectively). Thus, DC-CIK cell immunotherapy prolonged the OS of patients with advanced cancer.

The association between the T-cell subsets and the clinical outcome of advanced cancer was also investigated, as any successful host immune response against a tumor requires a well-balanced positive and negative regulation of lymphocytes. Imbalance of the host cellular immunity may trigger cancer progression disease progression and treatment failure (28). Previous studies have demonstrated significant changes of peripheral blood lymphocyte cell subsets in patients with different malignant lesions and poor prognosis (2932). Consequently, we also investigated the T-cell subsets before and after treatment in both groups, which indicated that DC-CIK infusions altered the ratios of T-cell subsets, increasing the T-helper and cytotoxic T lymphocyte (CTL) subsets, while decreasing regulatory T lymphocyte (Treg) subsets, particularly the CD8+/CD28 subset.

Emerging evidence supports the hypothesis that the improvement of the host immune status, including the anti-PD-1 antibody and anti-CTLA4 antibody, may favor the clinical outcome of cancer patients (33). Our previous data also indicated that elevated levels of CD8+/CD28 suppressor T lymphocytes represent a novel independent predictor of progression-free survival in patients with metastatic breast cancer during post-chemotherapy follow-up (31). The present study revealed that T-cell subsets significantly affected the OS of patients with advanced cancer, and that DC-CIK treatment did not only improve the imbalance in the immune status, but also prolonged the OS in advanced cancer patients.

According to these results, there was a significant difference in the OS among the three groups (no immunotherapy, 1 cycle and ≥2 cycles of DC-CIK infusion). One cycle of DC-CIK infusion significantly altered the T-cell subsets, but no obvious difference was observed among the groups receiving 2, 3 and ≥4 cycles of DC-CIK infusion, which was likely due to the small number of cases. A larger randomized clinical trial will be conducted in the near future to confirm the treatment benefit and the preferred treatment courses of DC-CIK cells for patients with advanced cancer.

No serious adverse events were observed in the patients receiving DC-CIK cell immunotherapy, which was consistent with the results of other studies (22). Therefore, DC-CIK cells are able to eliminate tumor cells without severe injury to normal tissues, which renders this treatment suitable for elderly and advanced-stage cancer patients.

It should be noted that there were several limitations to this study. First, the results were generated from a retrospective observational study, and a prospective paired study is required to confirm the clinical outcomes of DC-CIK cell immunotherapy. Second, only 90 patients were considered eligible for the present study. More cases are required and randomized clinical studies according to different tumor types must be performed to further analyze the treatment benefits of DC-CIK cells for patients with advanced cancer. Third, it must be mentioned that the CD4+/CD25+ T-cell subsets detected in this study may not represent CD4+/CD25+FoxP3+ Tregs, as not only Tregs but different T-cell subsets were considered. Foxp3 expression will be included along with CD4 and CD25 to validate the reduction of Tregs in a following study.

The data presented herein demonstrated that T-cell subset changes were associated with the OS of advanced cancer patients, while DC-CIK cell immunotherapy may regulate and enhance the host's immune function, significantly improving the OS of patients with advanced cancer. No severe side effects were recorded during the immunotherapy process, indicating that this is a safe treatment modality. A larger randomized, prospective clinical trial is required to further validate the clinical efficacy of DC-CIK cell therapy for patients with advanced cancer, and to elucidate the detailed underlying mechanism.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81470139).

References

1 

Stewart BW and Wild CP: World Cancer Report 2014International Agency for Research on Cancer. Lyon: World Health Organization; 2014, View Article : Google Scholar

2 

Abrams J, Conley B, Mooney M, Zwiebel J, Chen A, Welch JJ, Takebe N, Malik S, McShane L, Korn E, et al: National cancer institute's precision medicine initiatives for the new national clinical trials network. Am Soc Clin Oncol Educ Book. 2014:71–76. 2014. View Article : Google Scholar

3 

Couzin-Frankel J: Breakthrough of the year 2013. Cancer immunotherapy. Science. 342:1432–1433. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Bernstein R: Translational research: Cancer killers. Nature. 508:139–140. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Sharma P and Allison JP: The future of immune checkpoint therapy. Science. 348:56–61. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Hontscha C, Borck Y, Zhou H, Messmer D and Schmidt-Wolf IG: Clinical trials on CIK cells: First report of the international registry on CIK cells (IRCC). J Cancer Res Clin Oncol. 137:305–310. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Stroncek D, Berlyne D, Fox B, Gee A, Heimfeld S, Lindblad R, Loper K, McKenna D Jr, Rooney C, Sabatino M, et al: Developments in clinical cell therapy. Cytotherapy. 12:425–428. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Zhang L, Wang J, Wei F, Wang K, Sun Q, Yang F, Jin H, Zheng Y, Zhao H, Wang L, et al: Profiling the dynamic expression of checkpoint molecules on cytokine-induced killer cells from non-small-cell lung cancer patients. Oncotarget. 7:43604–43615. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Rosenberg S: Lymphokine-activated killer cells: A new approach to immunotherapy of cancer. J Natl Cancer Inst. 75:595–603. 1985.PubMed/NCBI

10 

Rosenberg SA, Spiess P and Lafreniere R: A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 233:1318–1321. 1986. View Article : Google Scholar : PubMed/NCBI

11 

Yun YS, Hargrove ME and Ting CC: In vivo antitumor activity of anti-CD3-induced activated killer cells. Cancer Res. 49:4770–4774. 1989.PubMed/NCBI

12 

Yang L, Ren B, Li H, Yu J, Cao S, Hao X and Ren X: Enhanced antitumor effects of DC-activated CIKs to chemotherapy treatment in a single cohort of advanced non-small-cell lung cancer patients. Cancer Immunol Immunother. 62:65–73. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Liu J, Li H, Cao S, Zhang X, Yu J, Qi J, An X, Yu W and Ren X: Maintenance therapy with autologous cytokine-induced killer cells in patients with advanced epithelial ovarian cancer after first-line treatment. J Immunother. 37:115–122. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Tang X, Liu T, Zang X, Liu H, Wang D, Chen H and Zhang B: Adoptive cellular immunotherapy in metastatic renal cell carcinoma: A systematic review and meta-analysis. PLoS One. 8:e628472013. View Article : Google Scholar : PubMed/NCBI

15 

Luo H, Gong L, Zhu B, Huang Y, Tang C, Yu S, Yang Z and Zhou X: Therapeutic outcomes of autologous CIK cells as a maintenance therapy in the treatment of lung cancer patients: A retrospective study. Biomed Pharmacother. 84:987–993. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Kong DS, Nam DH, Kang SH, Lee JW, Chang JH, Kim JH, Lim YJ, Koh YC, Chung YG, Kim JM and Kim CH: Phase III randomized trial of autologous cytokine-induced killer cell immunotherapy for newly diagnosed glioblastoma in korea. Oncotarget. 8:7003–7013. 2017.PubMed/NCBI

17 

Li X, Dai X, Shi L, Jiang Y, Chen X, Chen L, Zhao J, Qiang W, Wu J, Ji M, et al: Phase II/III study of radiofrequency ablation combined with cytokine-induced killer cells treating colorectal liver metastases. Cell Physiol Biochem. 40:137–145. 2016. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Zhou C, Liu D, Li J, Sun H, Zheng X, Wang S, Hong G, Mallampati S, Sun H, Zhou X, et al: Chemotherapy plus dendritic cells co-cultured with cytokine-induced killer cells versus chemotherapy alone to treat advanced non-small-cell lung cancer: A meta-analysis. Oncotarget. 7:86500–86510. 2016.PubMed/NCBI

20 

Banchereau J and Steinman RM: Dendritic cells and the control of immunity. Nature. 392:245–252. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Gallimore A and Godkin A: Regulatory T cells and tumour immunity-observations in mice and men. Immunology. 123:157–163. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Lee JH, Lee JH, Lim YS, Yeon JE, Song TJ, Yu SJ, Gwak GY, Kim KM, Kim YJ, Lee JW and Yoon JH: Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology. 148:1383–1391.e6. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Zhao YJ, Jiang N, Song QK, Wu JP, Song YG, Zhang HM, Chen F, Zhou L, Wang XL, Zhou XN, et al: Continuous DC-CIK infusions restore CD8+ cellular immunity, physical activity and improve clinical efficacy in advanced cancer patients unresponsive to conventional treatments. Asian Pac J Cancer Prev. 16:2419–2423. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Li YC, Zhao L, Wu JP, Qu CX, Song QK and Wang RB: Cytokine-induced killer cell infusion combined with conventional treatments produced better prognosis for hepatocellular carcinoma patients with barcelona clinic liver cancer B or earlier stage: A systematic review and meta-analysis. Cytotherapy. 18:1525–1531. 2016. View Article : Google Scholar : PubMed/NCBI

25 

World Health Organization, . WHO Handbook for Reporting Results of Cancer Treatment. Geneva, Switzerland: WHO Offset publication No. 48; 1979

26 

Wu C, Jiang J, Shi L and Xu N: Prospective study of chemotherapy in combination with cytokine-induced killer cells in patients suffering from advanced non-small cell lung cancer. Anticancer Res. 28:3997–4002. 2008.PubMed/NCBI

27 

Sun S, Li XM, Li XD and Yang WS: Studies on inducing apoptosis effects and mechanism of CIK cells for MGC-803 gastric cancer cell lines. Cancer Biother Radiopharm. 20:173–180. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Zou W: Regulatory T cells, tumor immunity and immunotherapy. Nat Rev Immunol. 6:295–307. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Chen B, Liu L, Xu H, Yang Y, Zhang L and Zhang F: Effectiveness of immune therapy combined with chemotherapy on the immune function and recurrence rate of cervical cancer. Exp Ther Med. 9:1063–1067. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Wang ZX, Cao JX, Wang M, Li D, Cui YX, Zhang XY, Liu JL and Li JL: Adoptive cellular immunotherapy for the treatment of patients with breast cancer: A meta-analysis. Cytotherapy. 16:934–945. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Song G, Wang X, Jia J, Yuan Y, Wan F, Zhou X, Yang H, Ren J, Gu J and Lyerly HK: Elevated level of peripheral CD8(+)CD28(−) T lymphocytes are an independent predictor of progression-free survival in patients with metastatic breast cancer during the course of chemotherapy. Cancer Immunol Immunother. 62:1123–1130. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zhong R, Han B and Zhong H: A prospective study of the efficacy of a combination of autologous dendritic cells, cytokine-induced killer cells, and chemotherapy in advanced non-small cell lung cancer patients. Tumour Biol. 35:987–994. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Topalian SL, Drake CG and Pardoll DM: Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell. 27:450–461. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 7 Issue 5

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen F, Yang M, Song Q, Wu J, Wang X, Zhou X, Yuan Y, Song Y, Jiang N, Zhao Y, Zhao Y, et al: Enhanced antitumor effects and improved immune status of dendritic cell and cytokine‑induced killer cell infusion in advanced cancer patients. Mol Clin Oncol 7: 903-910, 2017
APA
Chen, F., Yang, M., Song, Q., Wu, J., Wang, X., Zhou, X. ... Ren, J. (2017). Enhanced antitumor effects and improved immune status of dendritic cell and cytokine‑induced killer cell infusion in advanced cancer patients. Molecular and Clinical Oncology, 7, 903-910. https://doi.org/10.3892/mco.2017.1415
MLA
Chen, F., Yang, M., Song, Q., Wu, J., Wang, X., Zhou, X., Yuan, Y., Song, Y., Jiang, N., Zhao, Y., Zhou, L., Ren, J."Enhanced antitumor effects and improved immune status of dendritic cell and cytokine‑induced killer cell infusion in advanced cancer patients". Molecular and Clinical Oncology 7.5 (2017): 903-910.
Chicago
Chen, F., Yang, M., Song, Q., Wu, J., Wang, X., Zhou, X., Yuan, Y., Song, Y., Jiang, N., Zhao, Y., Zhou, L., Ren, J."Enhanced antitumor effects and improved immune status of dendritic cell and cytokine‑induced killer cell infusion in advanced cancer patients". Molecular and Clinical Oncology 7, no. 5 (2017): 903-910. https://doi.org/10.3892/mco.2017.1415