3,401
Views
25
CrossRef citations to date
0
Altmetric
Review

Cell-based immunotherapy with cytokine-induced killer (CIK) cells: From preparation and testing to clinical application

, , , , , , , , , & show all
Pages 1379-1387 | Received 23 Sep 2016, Accepted 19 Jan 2017, Published online: 16 Mar 2017

ABSTRACT

Cell-based immunotherapy holds promise in the quest for the treatment of cancer, having potential synergy with surgery, chemotherapy and radiotherapy. As a novel approach for adoptive cell-based immunotherapy, cytokine-induced killer (CIK) cells have moved from the ‘bench to bedside’. CIK cells are a heterogeneous subset of ex-vitro expanded, polyclonal T-effector cells with both natural killer (NK) and T-cell properties, which present potent non-major histocompatibility complex-restricted cytotoxicity against a variety of tumor target cells. Initial clinical studies on CIK cell therapy have provided encouraging results and revealed synergistic antitumor effects when combined with standard therapeutic procedures. At the same time, issues such as inadequate quality control and quantity of CIK cells as well as exaggerated propaganda were continuously emerging. Thus, the Ministry of Health in China stopped CIK cell therapy in May 2016, which was a major setback for the innovation of CIK cell-based immunotherapy. Thus, it is very important to modify technical criteria to develop a standardized operation procedure (SOP) and standardized system for evaluating antitumor efficacy in a safe way.

Introduction

Cytokine-induced killer (CIK) cell therapy, as a new option in the field of cancer immunotherapy, is a set of therapeutic strategies based on manipulating and co-opting a patient's or close relative's cells to treat cancer.Citation1,2 CIK cells are in-vitro-expanded T lymphocytes characterized by the presence of 2 main subsets. The first subset with a CD3+CD56+ phenotype can exert wide mainly MHC-unrestricted antitumor activity against both solid and hematologic malignancies.Citation3-5 The second subset with a CD3+CD56 phenotype is more similar to conventional T lymphocytes.Citation6 Issues favoring their facile clinical translation are the ready availability of large amounts of expanded CIK cells, their MHC-unrestricted tumor killing, simple to grow, potential effectiveness against many tumor types, and low cost. CIK cells can migrate to the tumor site after intravenous infusion since they express several chemokine receptors.Citation7 CIK cells are only one among many immune cell-based therapies proposed for the treatment of cancer. Chimeric antigen receptor T (CART) cell therapy, another potential approach of immunotherapy, can recognize their target antigen via an interaction between the chimeric immuno-receptor and a cell surface ligand.Citation8 However, CART cell therapy is currently limited to hematologic malignancies and not widely applied due to its very high cost.Citation1,9 Numerous clinical trials have been conducted using CIK cells for the treatment of both hematological and solid tumors. However, the main problem to the use of CIK cell immunotherapy is the lack of fine precision in use of the cells. Intriguing future perspectives and challenges are under investigation for synergy with other immunotherapy approaches, targeted therapies or even conventional chemotherapy.Citation10-13

The WHO and RECIST criteria established since years have been used to assess the effectiveness of CIK cell-based therapy; however, these criteria are not sufficient to fully describe all the important processes and cytotoxic agents. Since 2016, the number of institutions working on immunotherapy in China has considerably increased. However, some of these institutions lacked sufficient magnitude of CIK cells and standard quality control procedures. Furthermore, the exaggerated commercialization of the CIK cell-based immunotherapy indirectly caused the death of a patient suffering from synovial sarcoma, owing to which the Ministry of Health in China decided to stop CIK cell-based immunotherapy until an appropriate criterion is set-up. An effective and standardized informed consent procedure for CIK cell-based immunotherapy is expected to reduce the risk involved. Meanwhile, continuous quality improvement (CQI) will help to detect and prevent errors, thereby reducing harm to patients.Citation14 Therefore, in the present study, the culture procedures for CIK cell-based immunotherapy, evaluation of cellular functions, and detection and management of contamination are presented. In addition, clinical examples involving cell-based tumor immunotherapy are discussed.

Growth and preparation of cells in Vitro

The brief preparation of CIK cells, as shown in , is based on the first clinical trial using cell-based cancer immunotherapy.Citation15 Although the procedures for CIK cell therapy lack quality control criteria, they have been relatively uniform, despite constant evolution in the past 30 y. The major procedures of CIK cell therapy followed on a large scale in Chinese cancer immunotherapy institutions are listed below and the corresponding operation is shown in .Citation16-18

Figure 2. The concentration of viable cells. All the data were randomized collected from 50 tumor patients of different classification at different stages. And the samples would be counted at least 3 times.

Figure 2. The concentration of viable cells. All the data were randomized collected from 50 tumor patients of different classification at different stages. And the samples would be counted at least 3 times.

Briefly, heparinized peripheral blood (50 ml) is collected from the patients with tumors. The peripheral blood mononuclear cells (PBMCs) are isolated by Ficoll density gradient (Tianjin HY, China) centrifugation, followed by washing with 0.9% clinical-grade saline solution thrice. The cells are then incubated at 37°C for 24 h, under 5% CO2, 95% relative humidity using X-VIVO 15 serum-free medium (Longza, Japan) containing 1000 U/ml IFN-γ (Peprotech, USA). After 24 h, anti-CD3 antibody (at a final concentration of 100 ng/ml, eBioscience, USA), 1 ng/ml IL-1α (Peprotech, USA) and 1000 U/ml recombinant human IL-2 (QuanGang, China) is added to the medium. After 5–7 d of culture, the final expansion levels are usually in the range of hundred fold and the cells are transferred into a clinical-grade cell culture bag. On days 7–10, cells are counted and 2.5 l of fresh medium containing 1000 U/ml rhIL-2 is added. On day 11, each of the products are sampled for sterility testing. Further, on day 14, autologous CIK cells are re-suspended in 200 ml normal saline and are confirmed to be free of bacterial, mycoplasma, or fungal contamination. The name and the admission number attached to the CIK cell bag are carefully checked before transfusion into the patient's body.

IFN-γ, added on day 0, plays its role through the activation of macrophages present in the culture, thus providing both contact-dependent and cytokine-mediated signals favoring the acquisition of a Th1 phenotype.Citation19-21

An initial mitogenic signal is provided by the anti-CD3 antibody (BD Biosciences, USA) and then sustained by the continuous presence of IL-1α and IL-2.Citation22,23

Testing of cells

Quantification of viable CIK cells

In modern biomedical research and in the bio-manufacturing industry, it is essential to routinely monitor the growth and proliferation of CIK cells by viable cell counting.Citation24 Every 2–4 days, the quality control personnel draws 3 ml of supernatant from the cell culture bag for cell counting to ensure the proper proliferation of CIK cells. All the samples are stained with 0.4% trypan blue (TB), and the cell size, concentration, and the viability of each sample is measured using the Countstar automated cell counter (Shanghai, China). At the end of the cultivation, the concentration of CIK cells should achieve 3–4 × 106/ml () and the viability should be > 92%.

Figure 1. The brief preparation of cytokine-induced killer (CIK) cells. (A) A brief description of CIK cells procedure in most clinical institutions on cell-based tumor immunotherapy. (B) The main preparation of CIK cells in lots of the immunotherapy institutions, which applied only about 50 ml whole blood cells instead of 100ml, and more monitoring measures were used during cultivation of the CIK cells. (NS, normal saline).

Figure 1. The brief preparation of cytokine-induced killer (CIK) cells. (A) A brief description of CIK cells procedure in most clinical institutions on cell-based tumor immunotherapy. (B) The main preparation of CIK cells in lots of the immunotherapy institutions, which applied only about 50 ml whole blood cells instead of 100ml, and more monitoring measures were used during cultivation of the CIK cells. (NS, normal saline).

Composition of CIK cells

The CIK cell population obtained is mainly a CD3+ heterogeneous T-cell population with 2 predominant subsets, positive (CD3+CD56+) and negative (CD3+CD56) for the simultaneous expression of CD56 membrane molecules, whereas the antitumor efficiency is mainly owing to the CD3+CD56+ cells.Citation25,26 The other cells, such as CD4+T cells, CD8+T cells, and T-regulatory lymphocytes (Treg cells) are mingled in the CIK cell population, which also influence the effect of immunotherapy.Citation27-30 On days 7, 10 and 14 of cultivation, the expressions of CD3, CD56, CD4, CD8, CD25, and CD127 molecules on the surface of CIK cells are detected by fluorescence-activated cell sorter (FACS, BD Bioscience, USA). The monoclonal antibodies used include anti- CD3+ to evaluate total T lymphocytes, CD3+/CD4+ to evaluate Helper T cells, CD3+/CD8+ to evaluate cytotoxic T cells (CTLs), CD3+/CD56+ to evaluate cytokine-induced killer cells (CIKs), CD3+/CD56- to evaluate natural killer (NK) cells, and CD4+/CD25+/CD127low/− to evaluate regulatory T cells (Tregs). Among them, the terminally differentiated CD3+CD56+ subset represents a key feature, a double T-cell and NK cell phenotype, which could exert a potent and widely MHC-unrestricted anti-tumor cytotoxicity. As a result, the percentage of CD3+CD56+ cells must exceed by 40%, with the proportion of CD8+T cells almost twice or more than the proportion of CD4+T cells. Simultaneously, the proportion of Tregs (CD4+CD25+CD127) within the CIK cell culture should also be significantly decreased, which suggests that the immunosuppressive function of Tregs has been inhibited (). Higher amount of CD3+CD56+ cells detected in the supernatants of culture results in more benefits to the tumor patients, such as improved prognosis, reduced recurrence, and prolonged recurrence-free span.

Figure 3. Immunological assessment of T-cell subgroup. The percentage of CD3+CD4+ cells, CD3+CD8+cells and CD3+CD56+cells were evaluated using 2-color FACS. Treg(CD3+CD56+CD127) cells were evaluated using 3-color FACS. Expression of CD25 and CD127 after gating on CD4+. The representative scatter plots were demonstrated here.

Figure 3. Immunological assessment of T-cell subgroup. The percentage of CD3+CD4+ cells, CD3+CD8+cells and CD3+CD56+cells were evaluated using 2-color FACS. Treg(CD3+CD56+CD127−) cells were evaluated using 3-color FACS. Expression of CD25 and CD127 after gating on CD4+. The representative scatter plots were demonstrated here.

Cytotoxicity of CIK cells against tumor cells

The CD3+CD56+ CIK cells derived from CD3+CD56 precursor T cells exhibit the highest cytotoxicity against target tumor cells.Citation31 The antitumor mechanisms of CIK cells include 2 signaling pathways: first, the binding of NK-cell receptors to their ligands.Citation32,33 and second, the induction of tumor cell apoptosis by Fas ligand via the Fas signaling pathway.Citation34 Therefore, on day 14 of cultivation, the cytotoxicity of CIK cells collected are estimated by co-culturing with K562, a CIK cell-sensitive target cell line, at effector/target (E/T) ratios of 1:1, 10:1, 20:1 and 40:1. The tumor-lysis efficiency is determined by using CCK-8 assay kits (Dojindo, Japan), and the major killing molecules, such as NKG2D, NK1.1, perforin, FasL, and granzyme B are monitored by FACS. The total cytolysis activity during cell culture has been used as a unique marker for the success of autologous CIK cell generation.

Cytokine secretion by CIK cells

Besides directly killing the tumor cells, CIK cells secrete numerous cytokines that induce antitumor immune responses in patients. Cytokines production by CIK cells is fast and efficient, leading to effective function of CIK cells. IFN-γ and TNF-α are the main cytokines produced by CIK cells,Citation35,36 which are involved in regulating innate and adaptive immunities. The other positive regulatory cytokines that could be detected in tumor immunotherapy laboratories are IL-2 and IL-4.Citation37-39 The negative regulatory cytokines detected are usually IL-10 and TGF-β.Citation40,41 CIK cells can be used as a vehicle for delivering biological cytokines directly to the tumor microenvironment. In addition, these cytokines participate in rebuilding the patient's immune defense following tumor excision, chemotherapy, or radiotherapy.Citation42

The aging of CIK cells

Aging in CIK cells is characterized by a decrease in genome integrity due to immune dysfunction and commensal dysbiosis. Telomeres are nucleoprotein structures located at the end of eukaryotic chromosomes. Overwhelming evidence has indicated that telomere shortening induces the instability of chromosomes, which could also contribute to exponential increases in genomic instability of adult cells in response to replicative aging.Citation43-45 Therefore, it is necessary to study aging during CIK cell proliferation and self-renewal by monitoring the activity or magnitude of telomeres on day 0, 7, and day 14.Citation46-49 Although there is no significant evidence to suggest the correlation between the efficiency of tumor immunotherapy and the age of CIK cells, understanding of this correlation is important for the effective development of CIK cell-based immunotherapy.

Microbiological contamination

Safety is the main prerequisite for the adoptive infusion of CIK cells in patients with malignant cancers.Citation50 During CIK cell transfusion, various agents may be transmitted. However, in the past 20 years, such a case was not reported in any immunotherapy laboratory owing to strict supervision and appropriate and prompt disposal strategies. Microbiological contamination such as bacteria, viruses, or endotoxins may enter during blood collection from a patient or during CIK cell cultivation. Before blood collection, the patient is subjected to a series of blood tests to screen for HIV, Hepatitis A/B/C virus, or Treponema pallidum (Syphilis). If the results of any of these tests are positive, all the processes involved in the patient's CIK cell extraction and cultivation are conducted in an isolated culture environment by specifically trained personnel. In addition, on day 11 of cell cultivation, the culture supernatants of all the CIK cells from different patients are collected for screening for bacteria, viruses, and endotoxins separately.Citation51-53

Autologous CIK cell transfusion

The autologous CIK cells are administered via intravenous infusion within a period of 30 min. The infusion of these cells is well-tolerated and does not cause severe adverse events such as fever, myalgia, flu-like symptoms, and fatigue.Citation54 In general, patients receive 4–8 cycles of CIK cell infusion in a year and if the situation remains stable, more cycles of CIK maintenance treatment are given using the protocol mentioned above. Conversely, the CIK therapy is stopped if the patients do not respond well.

Clinical use

A growing number of clinical trials have been performed and the outcome suggests that CIK therapy yields highly compelling clinical responses in several solid carcinomas, such as hepatocellular carcinoma or B-cell malignant lymphoma. The data of randomized clinical trials for CIK cell-based therapy were searched on PubMed and analyzed. Within the 68 matches found, 24 published papers were excluded due to the lack of clinical information and unrelated tumors and the clinical information from 44 papersCitation34,54-97 published on CIK cell therapy was analyzed (). The patients in the immunotherapy group received at least 4 cycles of transfusion of CIK-cells, with more than 1 × 109 CIK cells transfused into patients within 1 h via the peripheral vein.

Table 1. A summarization of the Clinical information on the CIK cell-based cancer immunotherapy.

The majority of tumor patients in these clinical trials were treated with CIK cells along with conventional therapy, to achieve a synergistic antitumor effect. Other studies suggest that the cytotoxicity of CIK cells can be enhanced and improved by DC vaccination and when transfected with IL-2 gene or activated by Retro Nectin, these CIK cells demonstrate a higher proliferation rate and efficient cytotoxic activity.Citation54,56,58-60,62-64,67,68,72,75,84 In general, all these published studies have demonstrated that CIK cell-based immunotherapy utilizes the body's natural ability to eliminate tumor cells by stimulating and restoring the ability of the immune system to recognize and kill tumor cells. In contrast to conventional therapeutic strategies, the application of CIK cells is associated with minimal side effects and no severe adverse events have been reported. Interestingly, a reduction of Hepatitis B virus load is observed in patients undergoing treatment with CIK cells in our laboratory and elsewhere also.Citation54

Owing to its easy availability and potent anti-tumor activity, CIK cell therapy is emerging as a promising immunotherapy approach in oncology and may improve the quality of life of tumor patients, when combined with standard therapeutic procedures.Citation98,99 However, the lack of large-scale clinical experiments and the standard period for immunotherapy are the main shortcomings. Currently, our institution is in the process of collecting huge amount of data from clinical experiments, with a hope to reveal new insights into the efficacy of the technique in future.

Conclusions

At present and in the immediate future, it appears that immunotherapy will hold a place second to standard cancer therapy, but not as an alternative.Citation100 CIK cell-based immunotherapy has demonstrated potential in improving clinical outcomes and relieving the major side effects of standard treatment options. Moreover, CIK cells in combination with other conventional and established therapeutic options represent an innovative approach and will probably provide a new insight into research in the future. In this article, we have discussed the complete SOP of CIK cell-based immunotherapy with a hope that a comprehensive criterion would be established as soon as possible. Furthermore, clinical trials suggest that the combination of conventional chemotherapy and CIK cell-based immunotherapy could represent a valuable option to overcome the first-line chemotherapy failure, which is frequently observed in cancer treatment.Citation74,90 Furthermore, the number of transfused CIK cells and frequency of transfusion seem to play an important role in enhancing antitumor activity.Citation28

However, there are also some limitations to our study. First, as the CIK cell-based therapy is restricted to Asian countries and doesn't involve clinical samples from different regions of the world, it fails to completely depict the effects of the treatment. Second, in most clinical trials, a limited number of patients are included, focusing only on a certain type of tumor. Third, the clinical data discussed here are selected from published papers rather than drawn directly from patient records. More large-scale clinical trials are required to confirm the role of CIK cell-based immunotherapy in cancer treatment. In conclusion, the combination of CIK cell-based immunotherapy and other constructive therapeutic procedures is likely to make a significant contribution to the treatment of patients with tumors.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgment

We thank Professor Fengping Shan for his professional suggestion and constructive comments on this manuscript.

Funding

This research work was supported by Doctoral Scientific Research Foundation of Liaoning Province (201601412 to Yiming Meng).

References

  • Schmidt TL, Negrin RS, Contag CH. A killer choice for cancer immunotherapy. Immunol Res 2014; 58:300-6; PMID:24791943; https://doi.org/10.1007/s12026-014-8507-2
  • Mesiano G, Todorovic M, Gammaitoni L, Leuci V, Giraudo Diego L, Carnevale-Schianca F, Fagioli F, Piacibello W, Aglietta M, Sangiolo D. Cytokine-induced killer (CIK) cells as feasible and effective adoptive immunotherapy for the treatment of solid tumors. Expert Opin Biol Ther 2012; 12(6):673-84; PMID:22500889; https://doi.org/10.1517/14712598.2012.675323
  • Elia A, Circosta P, SangiSSSolo D, Bonini C, Gammaitoni L, Mastaglio S, Genovese P, Geuna M, Avolio F, Inghirami G, et al. Cytokine-induced killer cells engineered with exogenous T-cell receptors directed against melanoma antigens: enhanced efficacy of effector cells endowed with a double mechanism of tumor recognition. Hum Gene Ther 2015; 26(4):220-31; PMID:25758764; https://doi.org/10.1089/hum.2014.112
  • Zanon C, Stocchero M, Albiero E, Castegnaro S, Chieregato K, Madeo D, Rodeghiero F, Astori G. Multivariate statistical data analysis as a tool to analyze ex vivo expansion dynamics of cytokine-induced killer cells. Cytometry B Clin Cytom 2014; 86(4):257-62; PMID:24002818; https://doi.org/10.1002/cyto.b.21124
  • Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med 1991; 174(1):139-49; PMID:1711560; https://doi.org/10.1084/jem.174.1.139
  • Lu PH, Negrin RS. A novel population of expanded human CD3+CD56+ cells derived from T cells with potent in vivo antitumor activity in mice with severe combined immunodeficiency. J Immunol 1994; 153(4):1687-96; PMID:7519209
  • Thorne SH, Negrin RS, Contag CH. Synergistic antitumor effects of immune cell-viral biotherapy. Science 2006; 311:1780-4; PMID:16556847; https://doi.org/10.1126/science.1121411
  • Ruella M, June CH. Chimeric antigen receptor T cells for B Cell Neoplasms: Choose the right CAR for you. Curr Hematol Malig Rep 2016; 11(5):368-84; PMID:27475429; https://doi.org/10.1007/s11899-016-0336-z
  • Guo Y, Wang Y, Han W. Chimeric antigen receptor-modified T cells for solid tumors: Challenges and prospects. J Immunol Res 2016; 2016:3850839; PMID:26998495
  • Nwangwu CA, Weiher H, Schmidt-Wolf IG. Increase of CIK cell efficacy by upregulating cell surface MICA and inhibition of NKG2D ligand shedding in multiple myeloma. Hematol Oncol 2016; PMID:27430430; https://doi.org/10.1002/hon.2326
  • Poh SL, Linn YC. Immune checkpoint inhibitors enhance cytotoxicity of cytokine-induced killer cells against human myeloid leukaemic blasts. Cancer Immunol Immunother 2016; 65(5):525-36; PMID:26961084; https://doi.org/10.1007/s00262-016-1815-8
  • Sangiolo D. Cytokine induced killer cells as promising immunotherapy for solid tumors. J Cancer 2011; 2:363-8; PMID:21716717; https://doi.org/10.7150/jca.2.363
  • Kaneko T, Fusauch Y, Kakui Y, Okumura K, Mizoguchi H, Oshimi K. Cytotoxicity of cytokine-induced killer cells coated with bispecific antibody against acute myeloid leukemia cells. Leuk Lymphoma 1994; 14(3-4):219-29; PMID:7950910; https://doi.org/10.3109/10428199409049672
  • Lee GR, Fitzgibbon MC, O'Shea P. Laboratory services: regaining and maintaining control. Int J Health Care Quality Assurance 2016; 29(5):507-22; PMID:27256774; https://doi.org/10.1108/IJHCQA-08-2015-0098
  • Schmidt-Wolf IG, Finke S, Trojaneck B, Denkena A, Lefterova P, Schwella N, Heuft HG, Prange G, Korte M, Takeya M, et al. Phase I clinical study applying autologous immunological effector cells transfected with the interleukin-2 gene in patients with metastatic renal cancer, colorectal cancer and lymphoma. Br J Cancer 1999; 81(6):1009-16; PMID: 10576658; https://doi.org/10.1038/sj.bjc.6690800
  • Wang QJ, Wang H, Pan K, Li YQ, Huang LX, Chen SP, He J, Ke ML, Zhao JJ, Li JJ, et al. Comparative study on anti-tumor immune response of autologous cytokine-induced killer (CIK) cells, dendritic cells-CIK (DC-CIK), and semi-allogeneic DC-CIK. Chin J Cancer 2010; 29(7):641-8; PMID:20591215; https://doi.org/10.5732/cjc.009.10772
  • Zhao JJ, Pan K, Wang QJ, Xu ZD, Weng DS, Li JJ, Li YQ, Xia JC. Effect of anti-asthma Chinese medicine Chuankezhi on the anti-tumor activity of cytokine-induced killer cells. Chin J Cancer 2013; 32(10):553-60; PMID:23470144
  • Chen CL, Pan QZ, Zhao JJ, Wang Y, Li YQ, Wang QJ, Pan K, Weng DS, Jiang SS, Tang Y, et al. PD-L1 expression as a predictive biomarker for cytokine-induced killer cell immunotherapy in patients with hepatocellular carcinoma. Oncoimmunology 2016; 5(7):e1176653; PMID:27622026; https://doi.org/10.1080/2162402X.2016.1176653
  • Hayes MP, Wang J, Norcross MA. Regulation of interleukin-12 expressionin human monocytes: selective priming by interferon-gamma of lipopolysaccharide-inducible p35 and p40 genes. Blood 1995; 86(2):646-50; PMID:7605994
  • Ma X, Chow JM, Gri G, Carra G, Gerosa F, Wolf SF, Dzialo R, Trinchieri G. The interleukin 12 p40 gene promoter is primed by interferon gamma in monocytic cells. J Exp Med 1996; 183(1):147-57; PMID:8551218; https://doi.org/10.1084/jem.183.1.147
  • Lopez RD, Waller EK, Lu PH, Negrin RS. CD58/LFA-3 and IL-12 provided by activated monocytes are critical in the in vitro expansion of CD56+ T cells. Cancer Immunol Immunother 2001; 49(12):629-40; PMID:11258789; https://doi.org/10.1007/s002620000148
  • Ochoa AC, Gromo G, Alter BJ, Sondel PM, Bach FH. Long-term growth of lymphokine-activated killer (LAK) cells: role of anti-CD3, beta-IL 1, interferon-gamma and -beta. J Immunol 1987; 138(8):2728-33; PMID:2435804
  • Anderson PM, Bach FH, Ochoa AC. Augmentation of cell number and LAK activity in peripheral blood mononuclear cells activated with anti-CD3 and interleukin-2. Preliminary results in children with acute lymphocytic leukemia and neuroblastoma. Cancer Immunol Immunother 1988; 27(1):82-8; PMID:3260824; https://doi.org/10.1007/BF00205763
  • Kim SI, Kim HJ, Lee HJ, Lee K, Hong D, Lim H, Cho K, Jung N, Yi YW. Application of a non-hazardous vital dye for cell counting with automated cell counters. Analytical Biochemistry 2015; 492(2016):8-12; PMID:26399556
  • Thanendrarajan S, Kim Y, Schmidt-Wolf I. New adoptive immunotherapy strategies for solid tumours with CIK cells. Expert Opin Biol Ther 2012; 12(5):565-72; PMID:22444075; https://doi.org/10.1517/14712598.2012.668879
  • Todorovic M, Mesiano G, Gammaitoni L, Leuci V, Giraudo Diego L, Cammarata C, Jordaney N, Carnevale-Schianca F, Gallo S, Fagioli F, et al. Ex vivo allogeneic stimulation significantly improves expansion of cytokine-induced killer cells without increasing their alloreactivity across HLA barriers. J Immunother 2012; 35(7):579-86; PMID:22892454; https://doi.org/10.1097/CJI.0b013e31826b1fd9
  • Ma Y, Zhang Z, Tang L, Xu YC, Xie ZM, Gu XF, Wang HX. Cytokine-induced killer cells in the treatment of patients with solid carcinomas: a systematic review and pooled analysis. Cytotherapy 2012; 14(4):483-93; PMID:22277010; https://doi.org/10.3109/14653249.2011.649185
  • Hirakawa M, Matos T, Liu H, Koreth J, Kim HT, Paul NE, Murase K, Whangbo J, Alho AC, Nikiforow S, et al. Low-dose IL-2 selectively activates subsets of CD4+Tregs and NK cells. JCI Insight 2016; 1(18):e89278; PMID:27812545; https://doi.org/10.1172/jci.insight.89278
  • Jiang J, Wu C, Lu B. Cytokine-induced killer cells promote antitumor immunity. J Transl Med 2013; 11(1):1-9; PMID:23281771; https://doi.org/10.1186/1479-5876-11-1
  • Cerundolo V, de la Salle H. Description of HLA class I- and CD8-deficient patients: Insights into the function of cytotoxic T lymphocytes and NK cells in host defense. Semin Immunol 2006; 18(6):330-6; PMID:16973375; https://doi.org/10.1016/j.smim.2006.07.006
  • Solana R, Casado JG, Delgado E, DelaRosa O, Marín J, Durán E, Pawelec G, Tarazona R. Lymphocyte activation in response to melanoma: interaction of NK-associated receptors and their ligands. Cancer Immunol Immunother 2007; 56(1):101-9; PMID:16485126; https://doi.org/10.1007/s00262-006-0141-y
  • Ghiringhelli F, Ménard C, Martin F, Zitvogel L. The role of regulatory T cells in the control of natural killer cells: relevance during tumor progression. Immunol Rev 2006; 214:229-38; PMID:17100888; https://doi.org/10.1111/j.1600-065X.2006.00445.x
  • Girardi M, Oppenheim DE, Steele CR, Lewis JM, Glusac E, Filler R, Hobby P, Sutton B, Tigelaar RE, Hayday AC. Regulation of cutaneous malignancy by gammadelta T cells. Science 2001; 294:605-9; PMID:11567106; https://doi.org/10.1126/science.1063916
  • Lettau M, Paulsen M, Schmidt H, Janssen O. Insights into the molecular regulation of FasL (CD178) biology. Eur J Cell Biol 2011; 90(6-7):456-66; PMID:21126798; https://doi.org/10.1016/j.ejcb.2010.10.006
  • Yang Y, Zhang J, Xia F, Zhang C, Qian Q, Zhi X, Yue C, Sun R, Cheng S, Fang S, et al. Human CIK cells loaded with Au Nanorods as a theranostic platform for targeted photoacoustic imaging and enhanced immunotherapy and photothermal therapy. Nanoscale Res Lett 2016; 11(1):285; PMID:27271853; https://doi.org/10.1186/s11671-016-1468-8
  • Kirkwood JM, Butterfield LH, Tarhini AA, Zarour H, Kalinski P, Ferrone S. Immunotherapy of cancer in 2012. CA Cancer J Clin 2012; 62(5):309-35; PMID:22576456; https://doi.org/10.3322/caac.20132
  • Linn YC, Hui KM. Cytokine-induced killer cells: NK-like T cells with cytotolytic specificity against leukemia. Leuk Lymphoma 2003; 44(9):1457-62; PMID:14565644; https://doi.org/10.1080/1042819031000083082
  • Loison E, Gougeon ML. Thimerosal compromises human dendritic cell maturation, IL-12 production, chemokine release, and T-helper polarization. Hum Vaccin Immunother 2014; 10(8):2328-35; PMID:25424939; https://doi.org/10.4161/hv.29520
  • Prinsen H, van Laarhoven HW, Pots JM, Duiveman-de Boer T, Mulder SF, van Herpen CM, Jacobs JF, Leer JW, Bleijenberg G, Stelma FF, et al. Humoral and cellular immune responses after influenza vaccination in patients with postcancer fatigue. Hum Vaccin Immunother 2015; 11(7):1634-40; PMID:25996472; https://doi.org/10.1080/21645515.2015.1040207
  • Zhu BQ, Ju SW, Shu YQ. CD137 enhances cytotoxicity of CD3+CD56+cells and their capacities to induce CD4+Th1 responses. Biomed Pharmacother 2009; 63(7):509-16; PMID:19019623; https://doi.org/10.1016/j.biopha.2008.10.003
  • Whiteside TL. Regulatory T cell subsets in human cancer: are they regulating for or against tumor progression? Cancer Immunol Immunother 2014; 63(1):67-72; PMID:24213679; https://doi.org/10.1007/s00262-013-1490-y
  • Weintraub K. The cancer defense. Sci Am 2016; 314(4):42-51; PMID:27082190; https://doi.org/10.1038/scientificamerican0416-42
  • Adams PD, Jasper H, Rudolph K. Aging-induced stem cell mutations as drivers for disease and cancer. Cell Stem Cell 2015; 16(2015):601-14; PMID:26046760; https://doi.org/10.1016/j.stem.2015.05.002
  • Barbé-Tuana FM, Parisi MM, Panizzutti BS, Fries GR, Grun LK, Guma FT, Kapczinski F, Berk M, Gama CS, Rosa AR. Shortened telomere length in bipolar disorder: a comparison of the early and late stages of disease. Rev Bras Psiquiatr 2016; 38(4):281-6; PMID:27798713
  • MacNeil DE, Bensoussan HJ, Autexier C. Telomerase regulation from beginning to the end. Genes (Basel) 2016; 7(9):pii: E64
  • Jaruga E. Activation of telomerase in a human tumor. Proc Natl Acad Sci U S A 1994; 91(8):2882-5; PMID:8159672; https://doi.org/10.1073/pnas.91.8.2882
  • Ahmed Z, Shah ZH, Rehman HM, Shahzad K, Daur I, Elfeel A, Hassan MU, Elsafori AK, Yang SH, Chung G. Genomics: A hallmark to monitor molecular and biochemical processes leading toward a better perceptive of seed aging and ex-situ conservation. Curr Issues Mol Biol 2016; 22:89-112; PMID:27801354
  • Brazvan B, Farahzadi R, Mohammadi SM, Montazer Saheb S, Shanehbandi D, Schmied L, Soleimani Rad J, Darabi M, Nozad Charoudeh H. Key Immune Cell Cytokines Affects the Telomere Activity of Cord Blood Cells In vitro. Adv Pharm Bull 2016; 6(2):153-61; PMID:27478776; https://doi.org/10.15171/apb.2016.022
  • Nguyen HH, Kim T, Song SY, Park S, Cho HH, Jung SH, Ahn JS, Kim HJ, Lee JJ, Kim HO, et al. Naïve CD8(+) T cell derived tumor-specific cytotoxic effectors as a potential remedy for overcoming TGF-β immunosuppression in the tumor microenvironment. Sci Rep 2016; 6:28208; PMID:27306834; https://doi.org/10.1038/srep28208
  • Su Y, Yang Y, Ma Y, Zhang Y, Rao W, Yang G, Kou C. The efficacy and safety of dendritic cells co-cultured with cytokine-induced killer cell therapy in combination with TACE-predominant minimally-invasive treatment for Hepatocellular Carcinoma: a meta-analysis. Clin Lab 2016; 62(4):599-608; PMID:27215078
  • Garcia RA, Spitzer ED, Beaudry J, Beck C, Diblasi R, Gilleeny-Blabac M, Haugaard C, Heuschneider S, Kranz BP, McLean K, et al. Multidisciplinary team review of best practices for collection and handling of blood cultures to determine effective interventions for increasing the yield of true-positive bacteremias, reducingcontamination, and eliminating false-positive central line-associated bloodstream infections. Am J Infect Control 2015; 43(11):1222-37; PMID:26298636; https://doi.org/10.1016/j.ajic.2015.06.030
  • Denno J, Gannon M. Practical steps to lower blood culture contamination rates in the emergency department. J Emerg Nurs 2013; 39(5):459-64; PMID:22727270
  • Snyder SR, Favoretto AM, Baetz RA, Derzon JH, Madison BM, Mass D, Shaw CS, Layfield CD, Christenson RH, Liebow EB. Effectiveness of practices to reduce blood culture contamination: a laboratory medicine best practices systematic review and meta-analysis. Clin Biochem 2012; 45(13-14):999-1011; PMID:22709932; https://doi.org/10.1016/j.clinbiochem.2012.06.007
  • Ma L, Wang Y, Bo J, Han W, Wang Y, Zhang L, Wu X, Yu S, Liu R. The efficacy and safety of dendritic cells co-cultured with Cytokine-Induced killer cell therapy in combination with TACE-predominant minimally-invasive treatment for Hepatocellular Carcinoma: a Meta-Analysis. Clin Exp Immunol 2016; 184(1):83-9; PMID:26660736; https://doi.org/10.1111/cei.12755
  • Wu C, Jiang J, Shi L, 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 2008; 28(6B):3997-4002; PMID:19192663
  • Li H, Wang C, Yu J, Cao S, Wei F, Zhang W, Han Y, Ren XB. Dendritic cell-activated cytokine-induced killer cells enhance the anti-tumor effect of chemotherapy on non-small cell lung cancer in patients after surgery. Cytotherapy 2009; 11(8):1076-83; PMID:19929470; https://doi.org/10.3109/14653240903121252
  • Li R, Wang C, Liu L, Du C, Cao S, Yu J, Wang SE, Hao X, Ren X, Li H. Autologous cytokine-induced killer cell immunotherapy in lung cancer: a phase II clinical study. Cancer Immunol Immunother 2012; 61(11):2125-33; PMID:22581306; https://doi.org/10.1007/s00262-012-1260-2
  • Yang L, Ren B, Li H, Yu J, Cao S, Hao X, 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 2013; 62(1):65-73; PMID:22744010; https://doi.org/10.1007/s00262-012-1311-8
  • Zhong R, Teng J, Han B, Zhong H. Dendritic cells combining with cytokine-induced killer cells synergize chemotherapy in patients with late-stage non-small cell lung cancer. Cancer Immunol Immunother 2011; 60(10):1497-502; PMID:21681372; https://doi.org/10.1007/s00262-011-1060-0
  • Zhao P, Bu X, Wei X, Sun W, Xie X, Li C, Guo Q, Zhu D, Wei X, Gao D. Dendritic cell immunotherapy combined with cytokine-induced killer cells promotes skewing toward Th2 cytokine profile in patients with metastatic non-small cell lung cancer. Int Immunopharmacol 2015; 25(2):450-6; PMID:25698555; https://doi.org/10.1016/j.intimp.2015.02.010
  • Jin CG, Chen XQ, Li J, Wu ZP, Liu X, Wang XC. Moderating effects and maintenance of lung cancer cellular immune functions by CIK cell therapy. Asian Pac J Cancer Prev 2013; 14(6):3587-92; PMID:23886150; https://doi.org/10.7314/APJCP.2013.14.6.3587
  • Zhong GC, Yan B, Sun Y, Zhang XY, Chen J, Su Y, Sun HP, Zhu B. Clinical efficacy of immunotherapy of dendritic cell and cytokine-induced killer cell combined with chemotherapy for treatment of multiple myeloma. Zhonghua Xue Ye Xue Za Zhi 2012; 33(12):1000-3; PMID:23363790
  • Zhao X, Ding HF, Xu M, Xing J, Lu G, Wang L, Liu GQ. Clinical Efficacy of dendritic cells and cytokine-induced killer cells combined with chemotherapy for treating newly diagnosed multiple myeloma and their effect on function of CD4(+) CD25(+) T Cells in Peripheral Blood. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2016; 24(1):122-6; PMID:26913407
  • Zhao X, Ji CY, Liu GQ, Ma DX, Ding HF, Xu M, Xing J. Immunomodulatory effect of DC/CIK combined with chemotherapy in multiple myeloma and the clinical efficacy. Int J Clin Exp Pathol 2015; 8(10):13146-55; PMID:26722513
  • Liang XF, Ma DC, Ding ZY, Liu ZZ, Guo F, Liu L, Yu HY, Han YL, Xie XD. Autologous cytokine-induced killer cells therapy on the quality of life of patients with breast cancer after adjuvant chemotherapy: a prospective study. Zhonghua Zhong Liu Za Zhi 2013; 35(10):764-8; PMID:24378099
  • Ren J, Di L, Song G, Yu J, Jia J, Zhu Y, Yan Y, Jiang H, Liang X, Che L, et al. Selections of appropriate regimen of high-dose chemotherapy combined with adoptive cellular therapy with dendritic and cytokine-induced killer cells improved progression-free and overall survival in patients with metastatic breast cancer: rearmament of such contentious therapeutic preferences. Clin Transl Oncol 2013; 15(10):780-8; PMID:23359185; https://doi.org/10.1007/s12094-013-1001-9
  • Wang H, Feng F, Zhu M, Wang R, Wang X, Wu Y, Zhuang Z. Therapeutic efficacy of dendritic cells pulsed by autologous tumor cell lysate in combination with CIK cells on advanced renal cell carcinoma. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2015; 31(1):67-71; PMID: 25575061
  • Wang D, Zhang B, Gao H, Ding G, Wu Q, Zhang J, Liao L, Chen H. Clinical research of genetically modified dendritic cells in combination with cytokine-induced killer cell treatment in advanced renal cancer. BMC Cancer 2014; 14:251; PMID:24720900; https://doi.org/10.1186/1471-2407-14-251
  • Liu L, Zhang W, Qi X, Li H, Yu J, Wei S, Hao X, Ren X. Randomized study of autologous cytokine-induced killer cell immunotherapy in metastatic renal carcinoma. Clin Cancer Res 2012; 18(6):1751-9; PMID:22275504; https://doi.org/10.1158/1078-0432.CCR-11-2442
  • Zhao X, Zhang Z, Li H, Huang J, Yang S, Xie T, Huang L, Yue D, Xu L, Wang L, et al. Cytokine induced killer cell-based immunotherapies in patients with different stages of renal cell carcinoma. Cancer Lett 2015; 362(2):192-8; PMID:25843292; https://doi.org/10.1016/j.canlet.2015.03.043
  • Zhang Y, Wang J, Wang Y, Lu XC, Fan H, Liu Y, Zhang Y, Feng KC, Zhang WY, Chen MX, et al. Autologous CIK cell immunotherapy in patients with renal cell carcinoma after radical nephrectomy. Clin Dev Immunol 2013; 2013:195691; PMID:24382970; https://doi.org/10.1155/2013/195691
  • Zhan HL, Gao X, Pu XY, Li W, Li ZJ, Zhou XF, Qiu JG. A randomized controlled trial of postoperative tumor lysate-pulsed dendritic cells and cytokine-induced killer cells immunotherapy in patients with localized and locally advanced renal cell carcinoma. Chin Med J (Engl) 2012; 125(21):3771-7; PMID:23106871
  • Wang H, Feng F, Zhu M, Wang R, Wang X, Wu Y, Zhuang Z. [Therapeutic efficacy of dendritic cells pulsed by autologous tumor cell lysate in combination with CIK cells on advanced renal cell carcinoma]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2015; 31(1):67-71; PMID: 25575061
  • Zhang J, Zhu L, Zhang Q, He X, Yin Y, Gu Y, Guo R, Lu K, Liu L, Liu P, et al. Effects of cytokine-induced killer cell treatment in colorectal cancer patients: a retrospective study. Biomed Pharmacother 2014; 68(6):715-20; PMID:25107840; https://doi.org/10.1016/j.biopha.2014.07.010
  • Zhu H, Yang X, Li J, Ren Y, Zhang T, Zhang C, Zhang J, Li J, Pang Y. Immune response, safety, and survival and quality of life outcomes for advanced colorectal cancer patients treated with dendritic cell vaccine and cytokine-induced killer cell therapy. Biomed Res Int 2014; 2014:603871; PMID:25136601
  • Shi L, Zhou Q, Wu J, Ji M, Li G, Jiang J, Wu C. Efficacy of adjuvant immunotherapy with cytokine-induced killer cells in patients with locally advanced gastric cancer. Cancer Immunol Immunother 2012; 61(12):2251-9; PMID:22674056; https://doi.org/10.1007/s00262-012-1289-2
  • Jiang J, Xu N, Wu C, Deng H, Lu M, Li M, Xu B, Wu J, Wang R, Xu J, et al. Treatment of advanced gastric cancer by chemotherapy combined with autologous cytokine-induced killer cells. Anticancer Res 2006; 26(3B):2237-42; PMID: 16821594
  • Zhao M, Wu PH, Zeng YX, Xia JC, Zhang FJ, Xian LJ, Zhang YP, Zhou K, Fan WJ, Zhang L, et al. Cytokine-induced killer cell fusion to lower recurrence of hepatocellular carcinoma after transcatheter arterial chemoembolization sequentially combined with radiofrequency ablation: a randomized trial. Zhonghua Yi Xue Za Zhi 2006; 86(26):1823-8; PMID:17054858
  • Wang JP, Li W, Huang ZL, Wu PH, Li XS, Wei YD, Zhou QM, Pan CC, Xia JC, Zhao M. Value of CIK in the treatment of TACE combined with RFA for HCC in long-term survival and prognostic analysis. Zhonghua Yi Xue Za Zhi 2012; 92(43):3062-6; PMID:23328379
  • Pan GZ, Xu JB, Hao L, Zhang J, Li SL, Yuan QZ. Effect of CIK on long-term survival in the treatment of HCC after RFA combined TACE. Zhonghua Yi Xue Za Zhi 2016; 96(9):689-92; PMID:27055505
  • Pan CC, Huang ZL, Li W, Zhao M, Zhou QM, Xia JC, Wu PH. Serum alpha-fetoprotein measurement in predicting clinical outcome related to autologous cytokine-induced killer cells in patients with hepatocellular carcinoma undergone minimally invasive therapy. Chin J Cancer 2010; 29(6):596-602; PMID:20507732; https://doi.org/10.5732/cjc.009.10580
  • Hao MZ, Lin HL, Chen Q, Ye YB, Chen QZ, Chen MS. Efficacy of transcatheter arterial chemoembolization combined with cytokine-induced killer cell therapy on hepatocellular carcinoma: a comparative study. Chin J Cancer 2010; 29(2):172-7; PMID:20109346; https://doi.org/10.5732/cjc.009.10410
  • Hui D, Qiang L, Jian W, Ti Z, Da-Lu K. A randomized, controlled trial of postoperative adjuvant cytokine-induced killer cells immunotherapy after radical resection of hepatocellular carcinoma. Dig Liver Dis 2009; 41(1):36-41; PMID:18818130; https://doi.org/10.1016/j.dld.2008.04.007
  • Guo W, Liu L, Wu D. Dendritic cell-cytokine induced killer cell immunotherapy combined with transcatheter arterial chemoembolization for hepatocellular carcinoma: safety and efficacy. Nan Fang Yi Ke Da Xue Xue Bao 2014; 34(5):674-8; PMID:24849434
  • Weng DS, Zhou J, Zhou QM, Zhao M, Wang QJ, Huang LX, Li YQ, Chen SP, Wu PH, Xia JC. Minimally invasive treatment combined with cytokine-induced killer cells therapy lower the short-term recurrence rates of hepatocellular carcinomas. J Immunother 2008; 31(1):63-71; PMID:18157013; https://doi.org/10.1097/CJI.0b013e31815a121b
  • Yu X, Zhao H, Liu L, Cao S, Ren B, Zhang N, An X, Yu J, Li H, Ren X. A randomized phase II study of autologous cytokine-induced killer cells in treatment of hepatocellular carcinoma. J Clin Immunol 2014; 34(2):194-203; PMID:24337625; https://doi.org/10.1007/s10875-013-9976-0
  • Zhou P, Liang P, Dong B, Yu X, Han Z, Xu Y. Phase clinical study of combination therapy with microwave ablation and cellular immunotherapy in hepatocellular carcinoma. Cancer Biol Ther 2011; 11(5):450-6; PMID:21258206; https://doi.org/10.4161/cbt.11.5.14669
  • Lee JH, Lim YS, Yeon JE, Song TJ, Yu SJ, Gwak GY, Kim KM, Kim YJ, Lee JW, Yoon JH. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology 2015; 148(7):1383-91; PMID:25747273; https://doi.org/10.1053/j.gastro.2015.02.055
  • Shi M, Zhang B, Tang ZR, Lei ZY, Wang HF, Feng YY, Fan ZP, Xu DP, Wang FS. Autologous cytokine-induced killer cell therapy in clinical trial phase I is safe in patients with primary hepatocellular carcinoma. World J Gastroenterol 2004; 10(8):1146-51; PMID:15069715
  • Li W, Wang Y, Zhao L, Xu L, Zhang Y, Mai L, Gao Q. Efficacy of RetroNectin-activated cytokine-induced killer cell therapy in metastatic brain tumor patients. Oncol Res Treat 2015; 38(4):160-5; PMID:25877939; https://doi.org/10.1159/000380890
  • Olioso P, Giancola R, Di Riti M, Contento A, Accorsi P, Iacone A. Immunotherapy with cytokine induced killer cells in solid and hematopoietic tumours: a pilot clinical trial. Hematol Oncol 2009; 27(3):130-9; PMID:19294626; https://doi.org/10.1002/hon.886
  • Lu XC, Yang B, Yu RL, Chi XH, Tuo S, Tuo CW, Zhu HL, Wang Y, Jiang CG, Fu XB, et al. Clinical study of autologous cytokine-induced killer cells for the treatment of elderly patients with diffuse large B-cell lymphoma. Cell Biochem Biophys 2012; 62(1):257-65; PMID:21913005; https://doi.org/10.1007/s12013-011-9273-6
  • Wang H, Cao F, Li J, Li Y, Liu X, Wang L, Liu Z, Li Y, Zhao H, Zhou J. Homing of cytokine-induced killer cells during the treatment of acute promyelocytic leukemia. Int J Hematol 2014; 100(2):165-70; PMID:24929848; https://doi.org/10.1007/s12185-014-1618-7
  • Wang M, Shi SB, Qi JL, Tang XY, Tian J. S-1 plus CIK as second-line treatment for advanced pancreatic cancer. Med Oncol 2013; 30(4):747; PMID:24122257; https://doi.org/10.1007/s12032-013-0747-9
  • Chung MJ, Park JY, Bang S, Park SW, Song SY. Phase II clinical trial of ex vivo-expanded cytokine-induced killer cells therapy in advanced pancreatic cancer. Cancer Immunol Immunother 2014; 63(9):939-46; PMID:24916038; https://doi.org/10.1007/s00262-014-1566-3
  • Yan L, Wu M, Ba N, Wang LJ, Zhang HQ, Shi GY, Zhang ZS, Wang XJ. Efficacy of dendritic cell-cytokine-induced killer immunotherapy plus intensity-modulated radiation therapy in treating elderly patients with esophageal carcinoma. Genet Mol Res 2015; 14(1):898-905; PMID:25730028; https://doi.org/10.4238/2015.February.2.13
  • Li Y, Pan K, Liu LZ, Li YQ, Gu MF, Zhang H, Shen WX, Xia JC, Li JJ. Sequential cytokine-induced killer cell immunotherapy Enhances the efficacy of the gemcitabine plus cisplatin chemotherapy regimen for metastatic nasopharyngeal carcinoma. PLoS One 2015; 10(6):e0130620; PMID:26098948; https://doi.org/10.1371/journal.pone.0130620
  • Hontscha C, Borck Y, Zhou H, Messmer D, Schmidt-Wolf IG. Clinical trials on CIK cells: first report of the international registry on CIK cells (IRCC). J Cancer Res Clin Oncol 2011; 137(2):305-10; PMID:20407789; https://doi.org/10.1007/s00432-010-0887-7
  • Schmeel LC, Schmeel FC, Coch C, 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 2015; 141(5):839-49; PMID:25381063; https://doi.org/10.1007/s00432-014-1864-3
  • Guo Y, Han W. Cytokine-induced killer (CIK) cells: from basic research to clinical translation. Chinese J Cancer 2015; 34(3):99-107; PMID:25962508; https://doi.org/10.1186/s40880-015-0002-1

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.