In vivo distribution and antitumor effect of infused immune cells in a gastric cancer model

  • Authors:
    • Xiaohui Du
    • Runsen Jin
    • Ning Ning
    • Li Li
    • Quansheng  Wang
    • Wentao Liang
    • Juchao Liu
    • Yingxin Xu
  • View Affiliations

  • Published online on: September 3, 2012     https://doi.org/10.3892/or.2012.2013
  • Pages: 1743-1749
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adoptive cellular transfer has been employed for cancer immunotherapy, including patients with gastric cancer. However, little is known about the distribution of effector cells after their injection via different pathways. In this study, we used human gastric cancer cells (BGC823) tagged with enhanced green fluorescent protein (EGPF) to establish a subcutaneous gastric cancer model in nude mice. Cytokine-induced killer (CIK) cells and cytotoxic T lymphocytes (CTLs) were generated from human peripheral blood and labeled with red fluorescent PKH26. A portion of CIK cells was armed with CEA/CD3-bispecific single-chain antibody. When CIK cells were injected into nude mice with established subcutaneous gastric cancer via peritumoral (p.t.), intravenous (i.v.) and intraperitoneal (i.p.) infusion respectively, the distribution of cells was observed using a live fluorescence imaging system. We found that only a very small number of CIK cells could travel to the tumor site after i.p. or i.v. infusion, and they inhibited subcutaneous tumor growth in vivo only immediately following injection. In contrast, p.t. injection resulted in a significantly higher accumulation of CIK cells at the tumor site for 48 hours and mediated the greatest tumor inhibition compared with the other two injection methods. In addition, we compared the antitumor activity of CIK, CEA/CD3-bscAb-CIK and CTL cells in vitro and in vivo after p.t. injection. Among the three types of immune cells, CTLs demonstrated the strongest antitumor activity both in vitro and in vivo. CEA/CD3-bispecific single chain antibody could effectively link T lymphocytes and tumor cells expressing CEA, and resulted in significantly higher accumulation of CIK cells at the tumor site compared with the parental CIK cells. This study indicates that peritumoral injection of immune effector cells by minimally invasive surgical procedures represents an effective delivery method of adoptive cellular immunotherapy. Tumor-specific immune cells, such as CTLs, are a better choice of effector cells than CIKs in cellular immunotherapy. Furthermore, CD3+ immune cells armed with the CEA/CD3-bispecific single chain antibody could more effectively travel to and accumulate at the site of tumors expressing CEA, such as gastric cancer.

Introduction

Gastric cancer is the fourth most common cancer in the world (1). Standard methods of treatment, e.g. surgery, chemotherapy and radiotherapy have shown mixed success for early-stage patients. However, the prognosis of patients with advanced-stage gastric cancer is still extremely poor. Currently, transfusion of lymphocytes or adoptive cellular immunotherapy (ACI) have been used for the treatment of cancer both in animal studies (24) and clinic trials (57). ACI has also been used to treat gastric cancer with encouraging results, indicating that ACI can improve the prognosis of some patients, especially those who cannot tolerate surgery (8).

Cytokine-induced killer cells (CIKs) are commonly used as immune effector cells to treat cancer (911). The major component of the heterogeneous CIK cells, NKT cells, expresses both T cell marker CD3 and NK cell marker CD56 and exhibits non-major histocompatibility complex (MHC)-restricted cytotoxicity. This represents an effective mechanism in the treatment of cancer. Infusion of CIK cells is typically through an intravenous pathway; however, little is known regarding the number of cells that actually arrive at the tumor site with this method and how long these cells could survive in vivo.

Apart from CIK cells, many other immune cells such as lymphokine-activated killer (LAK) cells, tumor infiltrating lymphocytes (TILs) and antigen-specific cytotoxic T lymphocytes (CTLs) have been used in adoptive therapy (5). CTLs play a central role in antitumor immunity (12,13) and have demonstrated therapeutic efficacy of cancer immunotherapy both in vivo and in vitro. However, the in vivo distribution of these cells following various methods of injection is also unclear.

In this study, we infused PKH26-labeled human CIK cells or CTLs into nude mice with established EGFP-positive human gastric cancer through different pathways and sequentially examined the tissue distribution of CIK cells and CTLs using live fluorescence imaging. This study intended to identify the effective ACI cell type as well as the effective route of immune cell delivery to the tumor.

Materials and methods

Mice

Four-week-old female BALB/c nude (nu/nu) mice were acquired from the Animal Center of the Academy of Military Medical Sciences (Beijing, China). All animals were maintained in a pathogen-free environment and all animal protocols followed the experimental procedures of the National Institutes of Health Guide for Care and Use of Laboratory Animals.

Cell line

The poorly differentiated human gastric adenocarcinoma cell line, BGC823, was purchased from the Chinese Academy of Medical Sciences (Beijing, China). These cells are kept in our laboratory at the Institute of General Surgery, General Hospital of PLA (Beijing, China) and maintained in Dulbecco's modified Eagle's medium (DMEM) (Sigma, St. Louis, MO, USA).

DNA transfection and isolation of stable EGFP-expressing cells

BGC823 tumor cells were transfected with the pEGFP-C1 plasmid (Clontech, Mountain View, CA, USA) using the Xfect™ transfection reagent (Clontech). The neomycin resistance gene (neoR) in pEGFP-C1 plasmid allows stably transfected tumor cells to be selected using G418. After transfection, the cells were passaged at a ratio of 1:5 in selective medium that contained 200 μg/ml geneticin (G418; Sigma). The level of G418 was increased to 2,000 μg/ml in a stepwise manner (increased every day by 100 μg/ml). Cell clones expressing high levels of EGFP were isolated by limit dilution in 96-well plates. The EGFP-expressing clones were then amplified and transferred by conventional culture methods. This resulted in the identification of a cell line with bright EGFP fluorescence, designated as BGC823-EGFP that was used in this study.

Tumor challenge

Nude mice were challenged with 1×107 BGC823-EGFP cells, subcutaneously. Whole-body images were taken using the IVIS-200 Imaging System (Xenogen, Alameda, CA, USA) on Day 10.

Dendritic cell isolation and culture

Peripheral blood mononuclear cells (PBMCs) from healthy donor were cultured with Cellix-901 medium (Beijing XinMingLiTai Bio-technique, Co., Ltd., Beijing, China) for 2 h. Adherent cells were collected and further cultured in Cellix-901 medium with 1,000 IU/ml rhIL-4 (Cell Genix) and 1,000 IU/ml rhGM-CSF for 7 days to generate a dendritic cell (DC)-enriched cell population. DCs were then pulsed with 3 μl/ml Pseudomonas aeruginosa and 20 μg/ml BGC823 cell lysate followed by incubation for 12 h before use for the next step. To prepare BGC823 cell lysate, BGC823 cells were digested with trypsin, washed in normal saline and centrifuged at 450 × g for 5 min. The supernatant was then discarded and cells were resuspended in sterile water, and were frozen and thawed repeatedly 3 times. Lysate was obtained after centrifugation and filtration.

CIK, CTL and CIK-CEA/CD3-bscAb cell isolation and culture

Using a blood cell separator (Institute of Biomedical Engineering, Chinese Academy of Medical Sciences), 2–4×109 PBMCs from healthy donors were obtained. To prepare CIK, cell concentration was adjusted to 2×106 cells/ml in fresh, serum-free Cellix-601 medium (Beijing XinMingLiTai Bio-technique, Co., Ltd.) with 2,000 U/ml rhIFN-γ and incubated at 37°C in a humidified atmosphere of 50 ml/l CO2. After 24 h, anti-CD3 (50 ng/ml) and rhIL-2 (1,000 U/ml) were added. On Days 0, 4, 7 and 10 of culture, cell densities were determined and phenotypes were identified by flow cytometric analysis (FACS) (Becton-Dickinson, USA). To make CTL cells, PBMCs were incubated in tissue culture flasks with the Ag-pulsed DCs. To generate CIK-CEA/CD3-bscAb cells, CEA/CD3-bispecific single chain antibody (Beijing ABT Genetic Engineering Technology, Co., Ltd., Beijing, China) was added into the medium to bind CIK cells to obtain CIK-CEA/CD3-bscAb cells.

Immune cell labeling and in vitro assays

Immune cells were labeled with red fluorescent PKH26 (Sigma-Aldrich, Co., LLC.) following the kit instructions. CIK-, CIK-CEA/CD3-bscAb- and CTL-mediated cell cytotoxicity was evaluated using the lactate dehydrogenase (LDH) release assay according to the manufacturer's protocol. Cytotoxicity was assessed as [(Asample-Aspontaneous)/(Amaximum-Aspontaneous)] ×100%, (A, absorbance).

Tracing immune cells in vivo and evaluating the inhibition of tumor growth

Seven days after BGC823-EGFP cells were implanted into the nude mice, the tumor-bearing animals were separated into 3 groups. All mice were injected with 1×107 labeled CIKs, intraperitoneally (i.p., Group 1) ; intravenously (i.v., Group 2) or peritumorally (p.t., Group 3), respectively. The migration and distribution of the infused CIK cells were then observed using the IVIS-200 Imaging System (Xenogen) at 4, 24, 48 and 96 h after CIK cells injection. The lung, liver, spleen, kidney, stomach and intestine were collected on Days 2 and 8 for histopathological analysis. Tumor size and weight were assessed on Day 35 to evaluate the therapeutic efficacy of transferred CIK cells.

In separate experiments, we set up 3 groups to compare the therapeutic efficacy of CIK, CIK-CEA/CD3-bscAb and CTL cells. Each group of labeled immune cells was injected peritumorally at 1×107 cells/mouse. Migration and distribution of the infused immune cells were then observed at 4, 24, 48, 96 and 144 h after cell infusion.

Statistical analysis

Statistical analyses were performed with the SPSS 17.0 statistical software using the Student's t-test and ANOVA. P-values of <0.05 were considered statistically significant between the experimental groups.

Results

Antitumor effect of different immune cells in vitro

Firstly, we detected the antitumor effect of labeling on the immune cells. No difference was observed in cell morphology after labeling with PKH26. Strong red fluorescence was detected by flxuorescence microscopy (Fig. 1A) and FACS analysis showed that 99.37% of CIK cells were PKH26-positive (Fig. 1B). There was no significant difference in the antitumor activity between CIK and CIK-PKH26 cells (Table I).

Table I

In vitro antitumor effects of CIK cells before and after labeling with PKH26 (mean ± SD, n=3).

Table I

In vitro antitumor effects of CIK cells before and after labeling with PKH26 (mean ± SD, n=3).

5 h10 h


GroupsE/T ratioLDHCytotoxicity, %LDHCytotoxicity, %
CIK5:10.457±0.02370.51±3.610.555±0.02495.43±4.84
10:10.697±0.08184.44±4.480.894±0.090100
CIK-PKH265:10.460±0.01768.88±3.670.555±0.02393.78±3.06
10:10.701±0.06383.54±4.980.901±0.028100

[i] LDH, lactate dehydrogenase release assay; CIK, cytokine-induced killer cells.

In addition to CIK cells, CIK-CEA/CD3-bscAb cells and CTLs also showed strong antitumor activity against BGC823 cells in vitro. At E:T cell ratios of 5:1 and 10:1 and at 5 and 10 h, the BGC823 cell killing activity was as follows: CTL > CIK-CEA/CD3-bscAb > CIK cells (Table II).

Table II

Antitumor effect of different immune cells in vitro (mean ± SD, %, n=3).

Table II

Antitumor effect of different immune cells in vitro (mean ± SD, %, n=3).

5 h10 h


Groups5:1 E/T ratio10:1 E/T ratio5:1 E/T ratio10:1 E/T ratio
CIK70.51±3.6184.44±4.4895.43±4.84100
CIK-CEA/CD3-bscAb74.09±2.0188.01±2.88100c100
CTL85.59±5.14a,b97.47±2.20a,b100c100

a P<0.05 vs. the CIK group;

b P<0.05 vs. the CIK-CEA/CD3-bscAb group;

c P<0.01 vs. the CIK group. CIK, cytokine-induced killer cells; CTL, cytotoxic T lymphocytes.

Establishment of human gastric cancer (BGC823) in nude mice subcutaneously

Fluorescence imaging was employed to monitor the growth of human gastric cancer in nude mice. First of all, we successfully transfected BGC823 cells with EGFP (BGC823-EGFP cells) and selected them with G418 (Fig. 2A). We then injected parental BGC823 and BGC823-EGFP cells subcutaneously into the nude mice, respectively. The BGC823-EGFP subcutaneous tumor was visible with the IVIS-200 Imaging System within 10 days after tumor cell implantation (Fig. 2B).

EGFP transfection was found to have no effect on BGC823 cell morphology and cell proliferation in vitro (Fig. 2C) or tumor growth in vivo (Fig. 2D). In addition, no difference was observed in the histopathological analysis between BGC823 and BGC823-EGFP tumors (Fig. 2E and F).

Distribution and antitumor effect of CIK cells infused via three different pathways

When CIK cells were i.p. injected (Fig. 3A), red fluorescent PKH26-labeled CIK cells tended to gather first in the abdominal cavity. At 24 h post-injection, cells began to spread, and a small amount of accumulation could be observed in the tumor area before gradually dissipating. On Day 5 post-injection, the red fluorescence was nearly gone. In the i.v. group (Fig. 3B), CIK-PKH26 cells dispersed rapidly in the body, and a small amount of accumulation could be observed in the tumor area 24 h post-injection. On Day 5, the red fluorescence vanished. In contrast, in the peritumoral injection group (Fig. 3C), CIK-PKH26 cells gathered around the tumor after injection and dissipated very slowly. On Day 5, we could still detect a small amount of red fluorescence around the tumor.

On Day 2 post-injection via p.t. injection, labeled CIK cells were found to infiltrate the tumor bed in high numbers, although this infiltration was mainly localized around the tumor (Fig. 4A), while in the i.p. (Fig. 4B) and i.v. groups (Fig. 4C), the red fluorescent spots were much fewer. In the i.p. group, the labeled CIK cells were detected infiltrating multiple organs. The spleen was the most enriched in CIK cells (Fig. 4D), followed by the liver (Fig. 4E), kidney (Fig. 4F), intestine (Fig. 4G), stomach (Fig. 4H) and lung (Fig. 4I). On Day 8 post-injection, we found only a small quantity of red fluorescent spots in the liver and spleen in the i.p. and i.v. groups; however, in the peritumoral injection group, some labeled CIK cells were still found infiltrating the tumor bed (Fig. 4J), liver (Fig. 4K) and spleen (Fig. 4L).

The infusion of CIK cells via each of the three pathways, particularly peritumorally, was found to inhibit subcutaneous tumor growth in nude mice. Three weeks after CIK transfusion, tumors of the peritumoral injection group were significantly smaller than the normal saline (NS)-treated control group. Although not statistically significant, tumors of the i.p. and i.v. groups were also smaller. Four weeks after transfusion, tumor weight in the peritumoral, i.v. and i.p. injection groups was significantly lighter than the NS-treated control groups (Fig. 5).

Distribution and antitumor effect of different immune cells infused via peritumoral injection

Following peritumoral injection, all of the immune cells were gathered around the tumor. CIK cells were nearly gone 5 days post-injection (Fig. 6A), followed by the dispersal of CTL cells within 7 days (Fig. 6B). However, CIK-CEA/CD3-bscAb cells remained crowded around the tumor 7 days post-injection (Fig. 6C).

Peritumoral injection of three immune cell types, particularly CTLs, was found to inhibit subcutaneous tumor growth in nude mice (Fig. 7). Control tumor weight was 1.09±0.10 g 35 days after implantation. CTL, CIK-CEA/CD3-bscAb and CIK cells injected peritumorally 7 days after tumor implantation reduced tumor weight by 27.29±5.05, 20.41±8.35 and 18.58±6.01%, respectively.

Discussion

Whole-body imaging has proven to be a useful technology for the study of the dynamics of metastatic cancer. Green fluorescent protein (GFP) expression in cancer cells can help externally to image and follow the natural course or impediment of tumor progression and metastasis (14). Tumor motility, progression and metastasis can be visualized at the single-cell level in vivo with GFP (15). EGFP is the enhanced GFP, which is made for a heightened brightness (16). In this study, we have successfully established the stable EGFP-expressing human gastric adenocarcinoma BGC823 cells (BGC823-EGFP). These cells form subcutaneous tumors in nude mice. BGC823-EGFP cells have revealed identical biological characteristics as parental BGC823 cells, and can therefore be used to investigate tumor growth and metastasis (15,17,18).

CIK cells are polyclonal T effector cells generated when cultured under conditions of cytokine stimulation (19). The main effector cells of the heterogeneous CIK cell family express NK and T cell markers CD56 and CD3, respectively, and are referred to as NK-like T (NKT) cells. These cells possess non-MHC-restricted antitumor activity which means they do not require prior specific sensitization to induce the recognition of target cells (9,20,21). Over the years, CIK cells have been used for their antitumor activity against a variety of tumor targets (10,11,22). In this study, we found that CIK cells killed 100% of BGC823 cells within 10 h at the E:T cell ratio of 10:1 in vitro. CIK cells also exhibited antitumor activity in vivo. In our gastric cancer nude mouse model, infused CIKs inhibited subcutaneous tumor growth. Importantly, we observed that CIK cells infused via different pathways demonstrated different distribution and tumor inhibitory effect.

CIK cells are usually infused intravenously in adoptive cellular immunotherapy (ACI). Hazelrigg et al (23) found that the immune cells first arrived at the lungs after intravenous transfusion. In 2–6 h, accumulation in the lungs reached a peak and dissipated, with gradual accumulation in the liver, kidney and spleen. The overall cell distribution tended to stabilize within 24 h. Skitzki et al (24) reported that CIK cells could widely immigrate into most organs after intravenous transfusion; distribution was related to blood supply and immune properties of the organs as well as the order in which cells reached each organ. Furthermore, the in vivo cytotoxic activity of adoptively transferred immune cells is mainly observed in the initial peak period post-transfusion. These studies suggest that immune cells can effectively reach most organs and tumor tissue while maintaining their cytotoxic abilities. For leukemia, lymphoma and other non-solid tumors, CIK cells can spread throughout the body, including the bone marrow, via intravenous transfusion to kill tumor cells (25,26). However, for solid tumors, such as gastric cancer, the ability of CIKs to target and accumulate in tumor tissue remains to be fully defined.

In our study, we found that, after intravenous transfusion, CIK cells dissipated with the blood circulation and could indeed arrive at the tumor tissue. In comparison, intraperitoneally infused CIK cells first gathered in the abdominal cavity, then distribution followed the same course as the intravenous group by 24 h. In contrast, peritumoral injection resulted in the maintenance of CIK cells in the tumor tissue for the maximum amount of time examined. In this case migration of infused CIK cells to other organs, such as the liver and spleen, was evident but not to the degree of the i.v. or i.p. groups. In terms of tumor growth, peritumoral injection of CIK cells showed improved inhibition. Together, these results indicate that immune cells used for cancer adoptive immunotherapy, if injected directly into the tumor area, may be able to achieve the maximum antitumor effect.

In order to potentiate the antitumor activity of CIK cells, many immunological manipulations have been developed. For example stimulating factors such as Bacille Calmette-Guerin (27) and virus vaccine (28) were added in cell culture to improve cell proliferation. Interleukin-2 (IL-2) genes were transfected into CIK cells to enhance their IL-2 production and potentiate their cytotoxicity (29). DCs were engineered to present tumor antigens to CIK cells with the hopes of enhancing specific recognition of tumor cells and their subsequent killing (30,31). Furthermore, bispecific antibodies for CD3 on T and/or NKT cells and surface antigen on the target cells were used to promote the engagement of CIK cells with target cells (32,33). We have previously found that the gastric adenocarcinoma BGC823 cells express surface carcinoembryonic antigen (CEA). We therefore used CEA/CD3-bispecific single chain antibody to coat CIK cells. CEA/CD3-bscAb can pull together T lymphocytes and CEA-expressing tumor cells (34). In this study, CIK-CEA/CD3-bscAb cells showed stronger antitumor activity than CIK cells against BGC823 cells in vitro. Use of CEA/CD3-bscAb to arm CIK cells may represent a potential approach to enhance the antitumor activity of CIK cells.

CTL cells are a subpopulation of T cells with specific cytotoxicity. CD8+ CTLs are the most numerous members of the CTL subgroup. After priming by antigen presented by antigen presenting cells, CTLs can recognize and kill corresponding target cells (35,36). Adoptive transfer of antitumor CTLs has already been employed in clinical trials and has shown to be effective in adoptive immunotherapy of ovarian cancer, melanoma, breast cancer and renal carcinoma (12,3740). In this study, CTL cells were derived from PBMCs and stimulated by DCs loaded with BGC823 tumor cell lysate. We found that the antitumor activity against gastric cancer of the CTLs was much stronger than CIK cells not stimulated with Ag-loaded DCs.

In summary, our results indicate that immune cells such as CIKs and CTLs have strong and direct cytotoxic effects on gastric cancer cells, which suggests that immunotherapy using these immune cells, may become a novel treatment strategy for patients with gastric cancer following surgery, radiotherapy and chemotherapy.

Acknowledgements

This study was supported by the Natural Science Foundation of China (no. 60601018).

References

1 

Kamangar F, Dores GM and Anderson WF: Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol. 24:2137–2150. 2006. View Article : Google Scholar

2 

Kim HM, Kang JS, Lim J, et al: Antitumor activity of cytokine-induced killer cells in nude mouse xenograft model. Arch Pharm Res. 32:781–787. 2009. View Article : Google Scholar

3 

Klebanoff CA, Gattinoni L, Palmer DC, et al: Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice. Clin Cancer Res. 17:5343–5352. 2011.PubMed/NCBI

4 

Marcus A and Eshhar Z: Tumor-specific allogeneic cells for cancer therapy. Expert Opin Biol Ther. 11:1551–1554. 2011. View Article : Google Scholar : PubMed/NCBI

5 

June CH: Adoptive T cell therapy for cancer in the clinic. J Clin Invest. 117:1466–1476. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Di Stasi A, Tey SK, Dotti G, et al: Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 365:1673–1683. 2011.PubMed/NCBI

7 

Zhong JH, Ma L, Wu LC, et al: Adoptive immunotherapy for postoperative hepatocellular carcinoma: a systematic review. Int J Clin Pract. 66:21–27. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Toh U, Fujii T, Mishima M, et al: Conventional chemotherapy combined with the repetitive immune cell transfer for patients with refractory advanced gastric cancer. Gan To Kagaku Ryoho. 34:1931–1933. 2007.(In Japanese).

9 

Linn YC and Hui KM: Cytokine-induced NK-like T cells: from bench to bedside. J Biomed Biotechnol. 2010:4357452010.PubMed/NCBI

10 

Sangiolo D: Cytokine induced killer cells as promising immunotherapy for solid tumors. J Cancer. 2:363–368. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Ma Y, Zhang Z, Tang L, et al: Cytokine-induced killer cells in the treatment of patients with solid carcinomas: a systematic review and pooled analysis. Cytotherapy. 14:483–493. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Wright SE, Rewers-Felkins KA, Quinlin IS, et al: Cytotoxic T-lymphocyte immunotherapy for ovarian cancer: a pilot study. J Immunother. 35:196–204. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Hickey MJ, Malone CC, Erickson KE, et al: Implementing preclinical study findings to protocol design: translational studies with alloreactive CTL for gliomas. Am J Transl Res. 4:114–126. 2012.PubMed/NCBI

14 

Yang M, Baranov E, Jiang P, et al: Whole-body optical imaging of green fluorescent protein-expressing tumors and metastases. Proc Natl Acad Sci USA. 97:1206–1211. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Chishima T, Miyagi Y, Wang X, et al: Cancer invasion and micrometastasis visualized in live tissue by green fluorescent protein expression. Cancer Res. 57:2042–2047. 1997.PubMed/NCBI

16 

Heim R, Cubitt AB and Tsien RY: Improved green fluorescence. Nature. 373:663–664. 1995. View Article : Google Scholar : PubMed/NCBI

17 

Kaneko K, Yano M, Tsujinaka T, et al: Establishment of a visible peritoneal micrometastatic model from a gastric adenocarcinoma cell line by green fluorescent protein. Int J Oncol. 16:893–898. 2000.PubMed/NCBI

18 

Kaneko K, Yano M, Yamano T, et al: Detection of peritoneal micrometastases of gastric carcinoma with green fluorescent protein and carcinoembryonic antigen promoter. Cancer Res. 61:5570–5574. 2001.PubMed/NCBI

19 

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

20 

Linn YC, Lau LC and Hui KM: Generation of cytokine-induced killer cells from leukaemic samples with in vitro cytotoxicity against autologous and allogeneic leukaemic blasts. Br J Haematol. 116:78–86. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Lopez RD, Waller EK, Lu PH and 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. 49:629–640. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Niam M, Linn YC, Fook Chong S, et al: Clinical scale expansion of cytokine-induced killer cells is feasible from healthy donors and patients with acute and chronic myeloid leukemia at various stages of therapy. Exp Hematol. 39:897–903. 2011. View Article : Google Scholar

23 

Hazelrigg MR, Hirsch JI and Merchant RE: Distribution of adoptively transferred, tumor-sensitized lymphocytes in the glioma-bearing rat. J Neurooncol. 60:143–150. 2002. View Article : Google Scholar

24 

Skitzki J, Craig RA, Okuyama R, et al: Donor cell cycling, trafficking, and accumulation during adoptive immunotherapy for murine lung metastases. Cancer Res. 64:2183–2191. 2004. View Article : Google Scholar

25 

Edinger M, Cao YA, Verneris MR, Bachmann MH, Contag CH and Negrin RS: Revealing lymphoma growth and the efficacy of immune cell therapies using in vivo bioluminescence imaging. Blood. 101:640–648. 2003. View Article : Google Scholar

26 

Nishimura R, Baker J, Beilhack A, et al: In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood. 112:2563–2574. 2008. View Article : Google Scholar

27 

Brandau S and Bohle A: Activation of natural killer cells by Bacillus Calmette-Guerin. Eur Urol. 39:518–524. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Akagi J, Takai E, Tamori Y and Ogawa M: CD3+CD56+CD8+ cells demonstrating a suppressor T cell-like function in the peripheral blood of colon cancer patients. Int J Oncol. 19:561–566. 2001.

29 

Nagaraj S, Ziske C and Schmidt-Wolf IG: Human cytokine-induced killer cells have enhanced in vitro cytolytic activity via non-viral interleukin-2 gene transfer. Genet Vaccines Ther. 2:122004. View Article : Google Scholar : PubMed/NCBI

30 

Marten A, Renoth S, von Lilienfeld-Toal M, et al: Enhanced lytic activity of cytokine-induced killer cells against multiple myeloma cells after co-culture with idiotype-pulsed dendritic cells. Haematologica. 86:1029–1037. 2001.

31 

Ziske C, Marten A, Schottker B, et al: Resistance of pancreatic carcinoma cells is reversed by coculturing NK-like T cells with dendritic cells pulsed with tumor-derived RNA and CA 19-9. Mol Ther. 3:54–60. 2001. View Article : Google Scholar : PubMed/NCBI

32 

van Spriel AB, van Ojik HH and van De Winkel JG: Immunotherapeutic perspective for bispecific antibodies. Immunol Today. 21:391–397. 2000.PubMed/NCBI

33 

Flieger D, Kufer P, Beier I, Sauerbruch T and Schmidt-Wolf IG: A bispecific single-chain antibody directed against EpCAM/CD3 in combination with the cytokines interferon alpha and interleukin-2 efficiently retargets T and CD3+CD56+ natural-killer-like T lymphocytes to EpCAM-expressing tumor cells. Cancer Immunol Immunother. 49:441–448. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Lutterbuese R, Raum T, Kischel R, et al: Potent control of tumor growth by CEA/CD3-bispecific single-chain antibody constructs that are not competitively inhibited by soluble CEA. J Immunother. 32:341–352. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Savage P, Millrain M, Dimakou S, Stebbing J and Dyson J: Expansion of CD8+ cytotoxic T cells in vitro and in vivo using MHC class I tetramers. Tumour Biol. 28:70–76. 2007.

36 

Morishima N, Mizoguchi I, Okumura M, et al: A pivotal role for interleukin-27 in CD8+ T cell functions and generation of cytotoxic T lymphocytes. J Biomed Biotechnol. 2010:6054832010. View Article : Google Scholar : PubMed/NCBI

37 

Yee C, Thompson JA, Byrd D, et al: Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci USA. 99:16168–16173. 2002.PubMed/NCBI

38 

Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J and Andreesen R: Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J Clin Oncol. 24:5060–5069. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Butler MO, Lee JS, Ansen S, et al: Long-lived antitumor CD8+ lymphocytes for adoptive therapy generated using an artificial antigen-presenting cell. Clin Cancer Res. 13:1857–1867. 2007.PubMed/NCBI

40 

Yamaguchi Y, Ohshita A, Hironaka K, et al: Adoptive immunotherapy using autologous lymphocytes sensitized with HLA class I-matched allogeneic tumor cells. Oncol Rep. 16:165–169. 2006.

Related Articles

Journal Cover

November 2012
Volume 28 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Du X, Jin R, Ning N, Li L, Wang Q, Liang W, Liu J and Xu Y: In vivo distribution and antitumor effect of infused immune cells in a gastric cancer model. Oncol Rep 28: 1743-1749, 2012
APA
Du, X., Jin, R., Ning, N., Li, L., Wang, Q., Liang, W. ... Xu, Y. (2012). In vivo distribution and antitumor effect of infused immune cells in a gastric cancer model. Oncology Reports, 28, 1743-1749. https://doi.org/10.3892/or.2012.2013
MLA
Du, X., Jin, R., Ning, N., Li, L., Wang, Q., Liang, W., Liu, J., Xu, Y."In vivo distribution and antitumor effect of infused immune cells in a gastric cancer model". Oncology Reports 28.5 (2012): 1743-1749.
Chicago
Du, X., Jin, R., Ning, N., Li, L., Wang, Q., Liang, W., Liu, J., Xu, Y."In vivo distribution and antitumor effect of infused immune cells in a gastric cancer model". Oncology Reports 28, no. 5 (2012): 1743-1749. https://doi.org/10.3892/or.2012.2013