Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Oncotarget ; 7(13): 16248-61, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26919247

ABSTRACT

Bromodomain-containing protein 7 (BRD7) is a subunit of the PBAF complex, which functions as a transcriptional cofactor for the tumor suppressor protein p53. Down-regulation of BRD7 has been demonstrated in multiple types of cancer. This study aimed to investigate BRD7 expression and its tumor suppressive effect in hepatocellular carcinoma (HCC).The expression of BRD7 was examined in clinical specimens of primary HCC and in HCC cell lines through real-time quantitative PCR, western blot and immunohistochemistry. The prognostic value of BRD7 expression and its correlation with the clinicopathological features of HCC patients were statistically analyzed. The effect of BRD7 on the tumorigenicity of HCC was also examined using proliferation and colony-formation assays, cell-cycle assays, migration and cell-invasion assays, and xenograft nude mouse models. BRD7 was down-regulated in tumor tissues and HCC cell lines. BRD7 protein expression was strongly associated with clinical stage and tumor size. Kaplan-Meier survival curves revealed higher survival rates in patients with higher BRD7 expression levels compared to those with lower BRD7 levels. A multivariate analysis indicated that BRD7 expression was an independent prognostic marker. The re-introduction of BRD7 expression significantly inhibited proliferation, colony formation, migration and invasion and led to cell cycle arrest in HCC cells in vitro. Furthermore, experiments in mice suggested that BRD7 overexpression suppresses HCC tumorigenicity in vivo. In conclusions, our data indicated that BRD7 may serve as a tumor suppressor in HCC and may be a novel molecular target for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Chromosomal Proteins, Non-Histone/metabolism , Liver Neoplasms/pathology , Adult , Aged , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Female , Heterografts , Humans , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Mice , Mice, Nude , Middle Aged , Tumor Suppressor Proteins/metabolism
2.
Oncoimmunology ; 4(9): e1038017, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26405607

ABSTRACT

Postoperative non-small cell lung cancer (NSCLC) patients require adjuvant therapy to improve their prognosis. In this study, we investigated the efficacy of a sequential combination of autologous cellular immunotherapy (CIT) and chemotherapy for postoperative NSCLC. This retrospective study included 120 postoperative NSCLC patients: 60 cases received only chemotherapy; 33 cases received chemotherapy and sequential CIT with cytokine-induced killer (CIK) cells; and 27 cases received chemotherapy and sequential CIT with alternate CIK and natural killer (NK) cells. Survival analysis showed significantly higher overall survival rates in the CIT group compared with the control group. Overall survival was higher in patients who received CIT with alternate CIK and NK cells than those who received treatment with only CIK cells. Multivariate analysis showed that adjuvant CIT was an independent prognostic factor for overall survival of patients with NSCLC. In subgroup analyses, adjuvant CIT significantly improved the overall survival of patients with less than 60 y old and positive lymph node. In conclusions, these data indicate that adjuvant CIT, especially with alternate application of CIK and NK cells, is an effective therapeutic approach to prolong survival of patients with NSCLC, particularly for patients ≤60 y old with positive lymph nodes.

3.
Oncoimmunology ; 4(2): e976507, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25949875

ABSTRACT

Adoptive cell therapy (ACT) for cancers using autologous tumor-infiltrating lymphocytes (TILs) can induce immune responses and antitumor activity in metastatic melanoma patients. Here, we aimed to assess the safety and antitumor activity of ACT using expanded TILs following concurrent chemoradiotherapy (CCRT) in patients with locoregionally advanced nasopharyngeal carcinoma (NPC). Twenty-three newly diagnosed, locoregionally advanced NPC patients were enrolled, of whom 20 received a single-dose of TIL infusion following CCRT. All treated patients were assessed for toxicity, survival and clinical and immunologic responses. Correlations between immunological responses and treatment effectiveness were further studied. Only mild adverse events (AEs), including Grade 3 neutropenia (1/23, 5%) consistent with immune-related causes, were observed. Nineteen of 20 patients exhibited an objective antitumor response, and 18 patients displayed disease-free survival longer than 12 mo after ACT. A measurable plasma Epstein-Barr virus (EBV) load was detected in 14 patients at diagnosis, but a measurable EBV load was not found in patients after one week of ACT, and the plasma EBV load remained undetectable in 17 patients at 6 mo after ACT. Expansion and persistence of T cells specific for EBV antigens in peripheral blood following TIL therapy were observed in 13 patients. The apparent positive correlation between tumor regression and the expansion of T cells specific for EBV was further investigated in four patients. This study shows that NPC patients can tolerate ACT with TILs following CCRT and that this treatment results in sustained antitumor activity and anti-EBV immune responses. A larger phase II trial is in progress.

4.
Sci Rep ; 5: 9202, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25776856

ABSTRACT

The benefits of adjuvant cytokine-induced killer (CIK) cell immunotherapy for hepatocellular carcinoma (HCC) remain mixed among patients. Here, we constructed a prognostic nomogram to enable individualized predictions of survival benefit of adjuvant CIK cell treatment for HCC patients. Survival analysis showed that the median overall survival (OS) and progression-free survival (PFS) for patients in the hepatectomy/CIK combination group were 41 and 16 months, respectively, compared to 28 and 12 months for patients in the hepatectomy alone group (control). Based on multivariate analysis of the entire cohort, independent factors for OS were tumor size, tumor capsule, pathological grades, total bilirubin, albumin, prothrombin time, alpha-fetoprotein, and tumor number, which were incorporated into the nomogram. The survival prediction model performed well, as assessed by the c-index and calibration curve. Internal validation revealed a c-index of 0.698, which was significantly greater than the c-index value of the TNM (tumor-node-metastasis) staging systems of 0.634. The calibration curves fitted well. In conclusions, our developed nomogram resulted in more accurate individualized predictions of the survival benefit from adjuvant CIK cell treatment after hepatectomy. The model may provide valuable information to aid in the decision making regarding the application of adjuvant CIK cell immunotherapy.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Cytokine-Induced Killer Cells/immunology , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/mortality , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Liver Neoplasms/diagnosis , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Retrospective Studies , Treatment Outcome , Tumor Burden , Young Adult
5.
Clin Cancer Res ; 20(11): 3003-11, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24668644

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) is a high risk form of this disease, even after surgery, due to the absence of targets for hormone treatment and anti-Her-2 therapy. Chemotherapy is the main therapeutic strategy for such patients with breast cancer, although the outcome is often unsatisfactory. Thus, the development of combination adjuvant therapies is essential for improved prognosis in patients with TNBC. In this study, we investigated the efficacy of a sequential combination of cytokine-induced killer cell (CIK) infusion and chemotherapy for patients with post-mastectomy TNBC. EXPERIMENTAL DESIGN: From 2008 to 2012, 90 patients with post-mastectomy TNBC were included in this retrospective study: 45 cases received chemotherapy alone or with sequential radiotherapy; a further 45 cases received chemotherapy with/without radiotherapy and sequential CIK infusion. RESULTS: Survival analysis showed significantly higher disease-free survival (DFS) and overall survival (OS) rates in the CIK treatment group compared with the control group (P = 0.0382, P = 0.0046, respectively; log-rank test). Multivariate survival analysis showed that CIK adjuvant treatment was an independent prognostic factor for OS of patients with TNBC. In subgroup analyses, CIK adjuvant treatment significantly increased the DFS rate of patients with pathologic grade 3, and significantly increased the OS rate of patients in N1, N2, N3, IIB, III TNM (tumor-node-metastasis) stages, and with pathologic grade 3. CONCLUSIONS: These data indicate that adjuvant CIK treatment combined with chemotherapy is an effective therapeutic strategy to prevent disease recurrence and prolong survival of patients with TNBC, particularly those with lymph node metastasis, advanced TNM stage, and poor pathologic grade. Clin Cancer Res; 20(11); 3003-11. ©2014 AACR.


Subject(s)
Cytokine-Induced Killer Cells/immunology , Immunotherapy/methods , Triple Negative Breast Neoplasms/therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Chemotherapy, Adjuvant , Combined Modality Therapy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Mastectomy , Middle Aged , Prognosis , Proportional Hazards Models , Radiotherapy , Retrospective Studies , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
6.
Tumour Biol ; 35(1): 701-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23955802

ABSTRACT

Cytokine-induced killer (CIK) cells are ex vivo generated heterogeneous NK-like T lymphocytes. It is not very clear whether the phenotype of CIK cells is associated with their therapeutic efficacy to cancer patients. Thus, in this study, the association of phenotype of CIK cells and the overall survival of 121 patients with hepatocellular carcinoma (HCC), 74 patients with lung cancer and 42 patients with colorectal cancer, all of whom underwent surgical resection and received autogenous CIK cell therapy, was analyzed. We found that high ratio of the CD3+CD4+ subset was associated with poorer overall survival in colorectal cancer, but not HCC or lung cancer. A high ratio of the CD3+CD8+ subset was associated with improved overall survival in all three types of cancer. A high ratio of the CD3+CD56+ NK-like subset was associated with improved overall survival in lung and colorectal cancer, but not HCC. A high ratio of the CD3-CD56+ NK subset was associated with poorer overall survival in lung and colorectal cancer, but not HCC. In conclusion, the CD3+CD8+ and CD3+CD56+ subsets, especially the CD3+CD8+ subset, may be the major phenotypes responsible for anti-tumor immunity in vivo after autogenous CIK cell therapy.


Subject(s)
Cytokine-Induced Killer Cells/immunology , Cytokine-Induced Killer Cells/metabolism , Neoplasms/immunology , Neoplasms/mortality , Phenotype , Antigens, CD/metabolism , Female , Humans , Immunophenotyping , Immunotherapy, Adoptive , Male , Neoplasms/therapy
7.
Ann Surg Oncol ; 20(13): 4305-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23892527

ABSTRACT

BACKGROUND: Even after surgery, hepatocellular carcinoma (HCC) has poor prognosis; adjuvant therapy is needed to improve effectively the outcome of HCC patients. We evaluated the efficacy of cytokine-induced killer (CIK) cell infusion as an adjuvant therapy for postoperative HCC patients. METHODS: A total of 410 patients were studied retrospectively (January 2002 to January 2007): 206 received surgery alone; 204 received surgery and at least four cycles of CIK cell transfusion (CIK group). Kaplan-Meier and Cox regression analyses were used to explore differences in OS between two groups. RESULTS: The CIK group overall survival rates were significantly higher than that of the surgery-alone group (log-rank test; p = 0.0007). Multivariate survival analysis showed that CIK cell treatment was an independent prognostic factor. In subgroup analysis, patients who received ≥8 cycles of CIK cell transfusion exhibited significantly better survival than the <8 cycle group (p = 0.0272). There was no significant difference in overall survival in patients with ≤5-cm tumors between the CIK and surgery-alone groups (p = 0.7567). However, in patients with >5-cm tumors, the CIK group displayed significantly better overall survival than the surgery-alone group (p = 0.0002). CONCLUSIONS: Postoperative immunotherapy with CIK cell transfusion may be an effective adjuvant treatment for improving the outcomes of HCC patients; >8 cycles of CIK cell transfusion may ensure that patients derive maximal benefits. Moreover, patients with large tumors might benefit more from CIK cell adjuvant treatment than patients with small tumors.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cytokine-Induced Killer Cells/immunology , Hepatectomy , Immunotherapy , Liver Neoplasms/therapy , Neoplasm Recurrence, Local/prevention & control , Adjuvants, Immunologic , Adult , Aged , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Infusions, Intravenous , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Postoperative Care , Retrospective Studies , Survival Rate , Treatment Outcome , Young Adult
8.
PLoS One ; 7(7): e40364, 2012.
Article in English | MEDLINE | ID: mdl-22808142

ABSTRACT

BACKGROUND: The ARID1A gene encodes adenine-thymine (AT)-rich interactive domain-containing protein 1A, which participates in chromatin remodeling. ARID1A has been showed to function as a tumor suppressor in various cancer types. In the current study, we investigated the expression and prognosis value of ARID1A in primary gastric cancer. Meanwhile, the biological role of ARID1A was further investigated using cell model in vitro. METHODOLOGY/PRINCIPAL FINDINGS: To investigate the role of ARID1A gene in primary gastric cancer pathogenesis, real-time quantitative PCR and western blotting were used to examine the ARID1A expression in paired cancerous and noncancerous tissues. Results revealed decreased ARID1A mRNA (P = 0.0029) and protein (P = 0.0015) expression in most tumor-bearing tissues compared with the matched adjacent non-tumor tissues, and in gastric cancer cell lines. To further investigate the clinicopathological and prognostic roles of ARID1A expression, we performed immunohistochemical analyses of the 224 paraffin-embedded gastric cancer tissue blocks. Data revealed that the loss of ARID1A expression was significantly correlated with T stage (P = 0.001) and grade (P = 0.006). Consistent with these results, we found that loss of ARID1A expression was significantly correlated with poor survival in gastric cancer patients (P = 0.003). Cox regression analyses showed that ARID1A expression was an independent predictor of overall survival (P = 0.029). Furthermore, the functions of ARID1A in the proliferation and colony formation of gastric cell lines were analyzed by transfecting cells with full-length ARID1A expression vector or siRNA targeting ARID1A. Restoring ARID1A expression in gastric cancer cells significantly inhibited cell proliferation and colony formation. Silencing ARID1A expression in gastric epithelial cell line significantly enhanced cell growth rate. CONCLUSIONS/SIGNIFICANCE: Our data suggest that ARID1A may play an important role in gastric cancer and may serve as a valuable prognostic marker and potential target for gene therapy in the treatment of gastric cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Nuclear Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Aged , Blotting, Western , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Nuclear Proteins/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Transcription Factors/metabolism , Treatment Outcome , Tumor Stem Cell Assay
9.
Int J Biol Sci ; 7(1): 53-60, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21234303

ABSTRACT

In this study, we characterized the intratumoral expression of IL-17 and CD8(+) TILs in gastric adenocarcinoma patients after resection and determined the correlation between the survival probability of gastric adenocarcinoma patients and the expression of IL-17 in tumor. Expression of IL-17 and CD8 was assessed by immunohistochemistry, and the prognostic effects of intratumoral IL-17 expression and CD8(+) TILs were evaluated by Cox regression and Kaplan-Meier analysis. Immunohistochemical detection revealed the presence of IL-17 and CD8(+) cells in gastric adenocarcinoma tissue samples (90.6%, 174 out of 192 patients and 96.9%, 186 out of 192 patients, respectively). We have also found that intratumoral IL-17 expression was significantly correlated with age (p=0.004) and that the number of CD8(+)TILs was significantly correlated with UICC staging (p=0.012) and the depth of tumor invasion (p=0.022). The five-year overall survival probability among patients intratumorally expressing higher levels of IL-17 was significantly better than those expressing lower levels of IL-17 (p=0.036). Multivariate Cox proportional hazard analyses revealed that intratumoral IL-17 expression (HR: 0.521; 95% CI: 0.329-0.823; p=0.005) was an independent factor affecting the five-year overall survival probability. We conclude that low levels of intratumoral IL-17 expression may indicate poor prognosis in gastric adenocarcinoma patients.


Subject(s)
Adenocarcinoma/diagnosis , Interleukin-17/metabolism , Stomach Neoplasms/diagnosis , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , CD8 Antigens/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Predictive Value of Tests , Prognosis , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality
10.
Chin J Cancer ; 29(7): 641-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20591215

ABSTRACT

BACKGROUND AND OBJECTIVE: Cytokine-induced killer (CIK) cells and autologous dendritic cells-CIK (DC-CIK) cells co-cultured with autologous dendritic cells (DCs) and CIK cells are commonly used for immunotherapy recently. We compared the anti-tumor immune response of CIK cells, autologous DC-CIK cells, and semi-allogeneic DC-CIK cells to explore a more effective anti-tumor adoptive immunotherapy approach. METHODS: Peripheral monocytes were isolated from patients with renal carcinoma, lung cancer, or maxillary squamous cell carcinoma and their healthy adult children. Isolated cells were cultured and induced as DCs and CIK cells in vitro. CIK cells from patients were co-cultured with autologous DCs and DCs from their children respectively, generating DC-CIK cells and semi-allogeneic DC-CIK cells. The anti-tumor activities of autologous CIK cells, autologous DC-CIK cells, and semi-allogeneic DC-CIK cells were measured by LDH assay. Intracellular staining was used to test the secretion of cytokines. Flow cytometry was applied for detecting the phonotype changes of these three types of cells. Cell proliferation and cell apoptosis were detected by 5,6-carboxyfluorescein diacetate succinimidyl ester (CFSE) and Annexin V/PI respectively. RESULTS: Compared with autologous CIK cells and DC-CIK cells, semi-allogeneic DC-CIK cells significantly enhanced the anti-tumor activity and IFN-gamma secretion, reduced IL-4 secretion, increased the ratio of CD3(+)CD56(+) cells and CD3(+)CD8(+) cells, decreased the number of CD4(+)CD25(+) cells, promoted cell proliferation, and lessened cell apoptosis. CONCLUSIONS: Semi-allogeneic DC-CIK cells had a stronger anti-tumor effect than did autologous CIK cells and DC-CIK cells. Our results provided experimental evidence for clinical application of DC-CIK cells.


Subject(s)
Apoptosis , Cell Proliferation , Cytokine-Induced Killer Cells/immunology , Dendritic Cells/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytokine-Induced Killer Cells/cytology , Cytokine-Induced Killer Cells/metabolism , Cytokines/metabolism , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Dendritic Cells/metabolism , Hep G2 Cells , Humans , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Interleukin-4/metabolism , K562 Cells , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , L-Lactate Dehydrogenase/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Maxillary Neoplasms/metabolism , Maxillary Neoplasms/pathology
11.
Ai Zheng ; 27(7): 685-91, 2008 Jul.
Article in Chinese | MEDLINE | ID: mdl-18606059

ABSTRACT

BACKGROUND & OBJECTIVE: Suppressor of cytokine signaling 1 (SOCS1) plays a critical role in antitumor immunity. Down-regulating SOCS1 in antigen-presenting dendritic cells (DCs) could enhance antigen-specific antitumor immunity. This study was to investigate the antigen-specific antitumor effect and mechanism of DCs with siRNA-mediated inhibition of SOCS1, stimulated by OK-432 and pulsed with hepatocellular carcinoma cell line HepG2 antigens. METHODS: The expression of SOCS1 in immature DCs was down-regulated by RNA interference (RNAi). DCs were pulsed with lysate of HepG2 cells and stimulated with OK-432. The morphology of DCs was observed under converted phase microscopy. Phenotypic changes in cells after stimulation were characterized by flow cytometry (FCM). The Alamar Blue assay was adopted to evaluate the activation and proliferation of autologous lymphocytes induced by mature DCs. The cytotoxicity of cytotoxic T lymphocytes (CTLs) elicited by modified DCs to HepG2, EC109 and K562 cells was tested by the lactate dehydrogenase (LDH) assay. RESULTS: Cells displaying a typical morphology and phenotypic properties of mature DCs were obtained successfully. The expression of SOCS1 in DCs was down-regulated by SOCS1 RNAi. Mature DCs showed high expressions of CD80, CD83, CD86, and HLA-DR. Pulsing of DCs with lysate of HepG2 had no influence on the phenotypic properties of DCs. Down-regulating SOCS1 expression enhanced the maturation of DCs. The modified DC tumor vaccine stimulated the proliferation of autologous lymphocytes effectively, and the proliferation rate of T cells was (110.7+/-22.2)%. After being activated by modified DCs, TCLs exerted a specific and effective killing effect on HepG2 cells, but not on EC109 and K562 cells. CONCLUSION: Mature DCs could induce antigen-specific antitumor immunity against hepatocellular carcinoma after silencing of SOCS1 by siRNA, stimulation by OK-432 and pulsing of DCs with HepG2 cell antigens.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/immunology , Carcinoma, Hepatocellular/therapy , Dendritic Cells/immunology , Liver Neoplasms/therapy , Picibanil/pharmacology , RNA, Small Interfering/genetics , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Liver Neoplasms/immunology , Lymphocyte Activation , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology
12.
J Immunother ; 31(1): 63-71, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157013

ABSTRACT

The recurrence of hepatocellular carcinoma (HCC) after minimally invasive therapy is frequent. Adoptive immunotherapy is thought to be an effective method to lower recurrence and metastasis rates of malignant tumors. Therefore, 85 HCC patients after transcatheter arterial chemoembolization and radiofrequency ablation therapy were randomized to immunotherapy group and no adjuvant therapy group. Autologous cytokine-induced killer (CIK) cells were transfused via hepatic artery to the patients. The alteration of levels of lymphocyte subsets in peripheral blood of patients was examined by flow cytometry. All patients were screened by computed tomography every 2 months to observe the tumor recurrent conditions. After CIK cell infusions, the percentages of CD3+, CD4+, CD56+, CD3+CD56+ cells, and CD4+/CD8+ ratio increased from 68.6+/-11.0%, 31.1+/-9.0%, 15.6+/-7.9%, 5.2+/-3.1%, and 1.1+/-0.5 to 70.7+/-10.1%, 33.5+/-8.0%, 18.4+/-9.4%, 5.9+/-2.8%, and 1.3+/-0.7, respectively (P<0.05); whereas the percentage of CD8 cells decreased from 31.1+/-7.8% to 28.6+/-8.3% (P<0.05). The 1-year and 18-month recurrence rates of the study group were 8.9% and 15.6%, compared with 30.0% and 40.0% of the control group (both P value<0.05). The data suggest that CIK cell transfusion is an effective treatment. It can boost the immunologic function in HCC patients and plays an important role in reducing the recurrence rate of HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Immunotherapy, Adoptive/methods , Killer Cells, Lymphokine-Activated/transplantation , Liver Neoplasms/therapy , Lymphocyte Subsets/transplantation , Neoplasm Recurrence, Local/prevention & control , Adult , Aged , CD3 Complex/analysis , CD56 Antigen/analysis , CD8 Antigens/analysis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Combined Modality Therapy , Cytotoxicity Tests, Immunologic , Female , Humans , Immunophenotyping , Interferon-gamma/metabolism , Interleukin-4/metabolism , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/immunology , Liver Neoplasms/pathology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Middle Aged , Treatment Outcome
13.
Zhonghua Zhong Liu Za Zhi ; 29(6): 411-4, 2007 Jun.
Article in Chinese | MEDLINE | ID: mdl-17974271

ABSTRACT

OBJECTIVE: To investigate whether dendritic cells fused with tumor cells could elicit in vitro antitumor responses against renal cell carcinoma (RCC) cells. METHODS: Renal carcinoma cells were purified from tumor tissue excised from patients with metastatic RCC through tumor cell purifying technique and cultured in RPMI-1640 medium containing 10% FCS. Monocyte-derived DCs generated from peripheral blood mononuclear cell of RCC patients were cultured in the presence of human recombinant granulocyte-macrophage colony stimulating factor and interleukin-4. Tumor cells and DCs were cocultured in the presence of polyethylene glycol (PEG) to generate cell fusion. The phenotype of tumor cells, DCs and fusion cells were detected by flow cytometry. MTT was used to measure the ability of fusion cells to stimulate T cell proliferation. T cell-mediated antitumor responses were measured by lactate dehydrogenase release (LDH) assay for lysis of autologous tumor cells. RESULTS: The DCs expressed MHC class I, MHC class II and costimulatary molecules (CD80 and CD86), while the renal carcinoma cells expressed a high molecular glycoprotein MUC-1. The DC/tumor fusion cells coexpressed MUC-1 and the phenotype of DCs, and could stimulate T cell proliferation effectively. CTLs stimulated by the fusion vaccine showed distinct lytie activity in vitro to autologous tumor cells. CONCLUSION: Dendritic cells fused with tumor cells can elicit distinct antitumor responses in vitro against tumor cells from patients with metastatic RCC, providing a basis for further research on the clinical application of fusion vaccine in treatment for renal cancers.


Subject(s)
Cancer Vaccines/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Hybrid Cells/immunology , B7-2 Antigen/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Fusion , Cell Proliferation , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Hybrid Cells/cytology , Hybrid Cells/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mucin-1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
14.
Ai Zheng ; 26(7): 693-7, 2007 Jul.
Article in Chinese | MEDLINE | ID: mdl-17626742

ABSTRACT

BACKGROUND & OBJECTIVE: Mucin-1 (MUC1), a tumor-associated antigen, is an optional molecular target for antitumor immunotherapy protocols. This study was to establish a subcutaneous human esophageal cancer transplantation model with MUC1 high expression in nude mice that closely resembles the biological features of human esophageal cancer, and provide a in vivo model for MUC1-targeting immunotherapy of esophageal cancer. METHODS: Human esophageal carcinoma cell line EC-109 with MUC1 high expression was cultured, and subcutaneously transplanted into BALB/c athymic nude mice (4-5 weeks old). The growth status of transplanted tumor was observed. These subcutaneous tumors were examined histologically. The expression of proliferating cell nuclear antigen (PCNA) was detected by immunohistochemistry. Cell cycle and MUC1 expression of the transplanted tumor cells were analyzed by flow cytometry. RESULTS: Human esophageal carcinoma transplantation models with MUC1 high expression were successfully established in 6 of the 7 nude mice. The histological and biological characteristics of subcutaneous transplanted tumors were similar to those of human esophageal carcinoma. The mean PCNA label index of subcutaneous transplanted tumor cells was (63.5+/-3.6)%. The mean S-phase fraction of cell cycle was (37.6+/-3.7)%. The positive rate of MUC1 in subcutaneous transplanted tumor cells was 97.5%. CONCLUSION: Human esophageal carcinoma transplantation model with MUC1 high expression in nude mice is similar to human malignant tumors in biological characteristics, and can be used to investigate the biological characteristics of esophageal carcinoma, as well as provides an applicable animal model for research on MUC1-targeting immunotherapy of esophageal cancer.


Subject(s)
Disease Models, Animal , Esophageal Neoplasms/immunology , Mucin-1/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Esophageal Neoplasms/pathology , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Proliferating Cell Nuclear Antigen/metabolism
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 24(2): 189-91, 2007 Apr.
Article in Chinese | MEDLINE | ID: mdl-17407079

ABSTRACT

OBJECTIVE: To analyze the allelic loss of heterozygosity (LOH) in the region of chromosome 4p15.1-4q12 in nasopharyngeal carcinoma (NPC) patients with family history. METHODS: Tumor cells and lymphocytes were obtained from paraffin-embedded biopsied tissue section by microdissection. LOH detections were carried out on 25 NPC patients with family history by PCR-based microsatellite polymorphism analysis using 7 pairs of microsatellite markers primers. The microsatellite loci located in 4p15.1-4q12 region. Genescan software was used to analyse LOH at each locus. RESULTS: Ninety-two percent of NPC cases (23/25) with family history was showed at least one microsatellite marker of LOH. Higher frequencies of LOH were found at three loci: D4S238 (56%), D4S350 (50%), D4S1547 (50%). The minimal common region of deletion might be defined between D4S350 and D4S1547. CONCLUSION: The higher incidence of LOH at D4S350 and D4S1547 suggests that there may be a potential tumor suppressor gene located in the two regions.


Subject(s)
Chromosomes, Human, Pair 4/genetics , Loss of Heterozygosity , Nasopharyngeal Neoplasms/genetics , Adult , Family Health , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction
16.
Ai Zheng ; 25(11): 1414-8, 2006 Nov.
Article in Chinese | MEDLINE | ID: mdl-17094912

ABSTRACT

BACKGROUND & OBJECTIVE: Recently, micro-invasive treatments showed well application prospective in treating primary hepatocellular carcinoma (HCC), while tumor immunotherapy is a hot topic in tumor treatment. This study was to investigate the efficacy of cytokine-induced killer cells (CIK) combined micro-invasive treatments (transcatheter arterial chemoembolization and radiofrequency ablation) on the recurrence of HCC. METHODS: A total of 85 HCC patients without active lesions and metastasis, which were displayed by imaging examination after transcatheter arterial chemoembolization and radiofrequency ablation therapy, were divided randomly into 2 groups: 45 in study group, and 40 in control group. The patients in study group were transfused with CIK cells through hepatic artery after micro-invasive treatments, while the patients in control group were not. The levels of T lymphocyte subsets and native killer (NK) cells in peripheral blood of HCC patients before and after CIK cell transfusion were detected by flow cytometry (FCM). Tumor condition was observed by CT scanning every 2-3 months. RESULTS: The percentages of CD3+, CD4+, and CD56+ effect cells and the proportion of CD4+/CD8+ were increased from (68.6+/-11.0)%, (31.0+/-9.0)%, (15.6+/-7.8)%, and 1.1+/-0.5 to (70.7+/-10.1)% (P<0.05), (33.5+/-8.0)% (P<0.05), (18.4+/-9.4)% (P<0.05), and 1.3+/-0.6 (P<0.05) after CIK cell transfusion; while the percentages of CD8+ and CD3+ CD56+ cells were decreased from (31.1+/-7.8)% and (6.4+/-3.5)% to (28.6+/-8.3)% (P<0.05) and (5.2+/-3.3)% (P<0.05). The 1- and 1.5-year recurrence rates were 8.89% and 15.56% in study group, and 30.00% and 40.00% in control group (Chi(2) = 4.87 and 6.41, P <0.05). CONCLUSION: CIK cell transfusion after micro-invasive treatments may improve the immunologic function in HCC patients, and play an important role in reducing the recurrence rate of HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Killer Cells, Natural/transplantation , Liver Neoplasms/therapy , Neoplasm Recurrence, Local/prevention & control , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CD4-CD8 Ratio , Carcinoma, Hepatocellular/immunology , Catheter Ablation , Chemoembolization, Therapeutic , Combined Modality Therapy , Female , Humans , Interferon-gamma/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Liver Neoplasms/immunology , Lymphocyte Count , Lymphocyte Subsets/pathology , Male , Microtubule Proteins , Middle Aged , Saccharomyces cerevisiae Proteins
17.
World J Gastroenterol ; 12(15): 2437-40, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16688840

ABSTRACT

AIM: To define the common deleted region on the long arm of haman chromosome 7q linked to primary gastric carcinomas in Chinese by loss of heterozygosity (LOH) and its clinical significance. METHODS: Nine microsatellite markers distributed over chromosome 7q with an average marker density of 10cM were used to examine 70 primary gastric carcinomas for LOH by PCR amplification. The PCR products were separated by electrophoresis on polyacrylamide gel. Genescan and Genotyper softwares were used to analyze LOH. RESULTS: LOH with at least one marker on 7q occurred in 34.3% (12/50) of the tumors. Among them, LOH at D7S486 and D7S798 was higher in 24.0% (24/70) and 19.2% (5/26), respectively. By statistical analysis we also observed an obvious genotype-phenotype correlation on 7q (P<0.05). The frequency of LOH at D7S486 in patients with lymph node metastasis was significantly higher than that in those without lymph node metastasis (P=0.015). CONCLUSION: The high incidence of LOH at D7S486 and its correlation with poorer prognosis suggest that there might be putative tumor suppressor genes in this region involved in the tumorigenesis and progression of gastric carcinoma.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Stomach Neoplasms/genetics , Adult , Aged , Female , Genetic Linkage , Humans , Loss of Heterozygosity , Male , Microsatellite Repeats , Middle Aged
18.
Ai Zheng ; 25(5): 625-30, 2006 May.
Article in Chinese | MEDLINE | ID: mdl-16687087

ABSTRACT

BACKGROUND & OBJECTIVE: Nowadays, operation is the main treatment for renal cell carcinoma (RCC). But the prognosis of advanced RCC is poor because of its high recurrence rate and resistance to conventional treatments, such as chemotherapy and radiotherapy. Hence, novel and more effective therapeutic options for advanced RCC are needed. This study was to evaluate the clinical efficacy of autologous renal tumor lysate-loaded dendritic cells (DCs) in combination with cytokine-induced killer (CIK) cells on advanced RCC. METHODS: Peripheral blood mononuclear cells were isolated from 10 patients with advanced RCC, and cultured with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) to produce DCs. The DCs were pulsed with autologous renal tumor cell lysate. T lymphocytes were cultured with interferon-gamma (IFN-gamma), IL-2, CD3-moAb, and IL-1alpha to prepare CIKs. After nephrectomy, the patients received intradermal DC vaccination weekly for at least 8 times, and CIKs administration biweekly for at least 4 times. Clinical and immunologic responses were evaluated by imaging examination, T lymphocytes subset changes, and delayed-type hypersensitivity (DHT) reaction, respectively. RESULTS: During follow-up of 6-20 months (median, 11 months), 1 case of partial remission (PR), 2 cases of stable disease (SD), and 1 case of progressive disease (PD) were identified in the 4 patients with measurable diseases; 1 case of PD was identified in the 6 patients with no measurable diseases, 1 case was lost, and no progressive disease was identified. When treated for 2 months, the levels of CD3+, CD4+, CD4+/CD8+, CD56+ were increased significantly (P<0.05) as compared with those before treatment. DTH reaction was positive in 6 patients, including the patient with PR. Except transient fever and chill, no remarkable adverse event happened during or after the treatment. CONCLUSION: Autologous tumor cell lysate-pulsed DCs in combination with CIKs shows short-term efficacy on advanced RCC through inducing specific antitumor immunity, and the adverse events are tolerable.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Cytokine-Induced Killer Cells/immunology , Dendritic Cells/immunology , Kidney Neoplasms/therapy , Adult , Aged , Carcinoma, Renal Cell/immunology , Cytokines/blood , Female , Follow-Up Studies , Humans , Hypersensitivity, Delayed , Immunotherapy, Adoptive , Kidney Neoplasms/immunology , Male , Middle Aged , Remission Induction , Young Adult
19.
Cancer Genet Cytogenet ; 166(2): 166-72, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16631475

ABSTRACT

Gastric carcinoma is one of the most common malignancies in Asia. Although the allelic deletion of 7q has been reportedly associated with primary gastric carcinoma tumorigenesis, no predisposing genes in this region have been identified so far. Here, we report the results of genotype and loss of heterozygosity (LOH) analysis on 7q in this tumor. A panel of nine microsatellite markers distributed over the whole chromosome 7q was used for genotyping primary gastric carcinomas. Of 72 primary tumors LOH of D7S486 occurred in 24.0% (12/50) of cases. Fine mapping with 12 additional markers flanking D7S486 resulted in LOH of 30.36% (17/56) and defined one minimal deleted region in primary gastric carcinomas, a 90-kilobase region bounded by D7S2543 and D7S486 at 7q31.2. The allelic deletion correlates statistically with clinicopathologic variables. Our data suggest a possible link between putative tumor suppressor genes and gastric carcinoma in the 7q31 region.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Genes, Tumor Suppressor , Loss of Heterozygosity/genetics , Stomach Neoplasms/genetics , Adult , Aged , Chromosome Deletion , Female , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Physical Chromosome Mapping
20.
Ai Zheng ; 24(11): 1306-11, 2005 Nov.
Article in Chinese | MEDLINE | ID: mdl-16552953

ABSTRACT

BACKGROUND & OBJECTIVE: Previous studies have showed that chromosome 7q occurs a high frequency of loss of heterozygosity (LOH) in primary gastric carcinoma; D7S486 located in 7q31 is a locus with the highest frequency of LOH on 7q, which is significantly related to lymph node metastasis of tumor. Gastric carcinoma-related tumor suppressor genes (TSGs) might locate near the locus. In order to seek gastric carcinoma-related TSGs in a further narrow region, LOH of microsatellite marker loci on chromosome 7q31 was examined in primary gastric carcinoma to determine the minimal commonly deleted regions, and the possible roles of these regional deletions in gastric carcinoma pathogenesis were analyzed in this study. METHODS: Twelve microsatellite markers centering D7S486 locus on 7q31 region, with an average genetic distance of about 0.5 centimorgan (cM), were selected. DNAs were extracted from 78 specimens of gastric carcinoma and corresponding normal gastric mucosa tissues, amplified through multi-polymerase chain reaction (multi-PCR), and separated by electrophoresis. LOH of microsatellite marker loci was analyzed with GeneScan and Genotyper software to determine the minimal commonly deleted regions on 7q31. The possible roles of these regional deletions in gastric carcinoma pathogenesis were analyzed with consideration of clinicopathologic parameters. RESULTS: LOH of all 12 selected microsatellite marker loci was detected in primary gastric carcinoma, with the total frequency of 41.7% (40/72). The most frequent LOH was D7S486 locus (located in 7q31.2), with the frequency of 30.4% (17/56); the second was D7S650 locus (located in 7q31.3), with the frequency of 21.1% (8/38). Two minimal commonly deleted regions, D7S2543-D7S486 and D7S480-D7S650 (each span about 90 kb), were detected on 7q31 in primary gastric carcinoma. The LOH frequency of D7S2543-D7S486 region was significantly related to clinical stage and lymph node metastasis (P = 0.01, P = 0.03); the LOH frequency of D7S480-D7S650 region was significantly related to clinical stage (P = 0.03), and LOH of this region occurred only in the patients with stage III/IV tumor, T3/T4 primary lesion, or lymph node metastasis. CONCLUSIONS: There are 2 minimal commonly deleted regions on 7q31 in primary gastric crcinoma, D7S2543-D7S486 and D7S480-D7S650. There might be TSGs in these 2 regions closely related to the progression of gastric carcinoma.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Genes, Tumor Suppressor , Loss of Heterozygosity/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , DNA, Neoplasm/genetics , Female , Humans , Lymphatic Metastasis , Male , Microsatellite Repeats , Middle Aged , Neoplasm Staging , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...