Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Oncotarget ; 8(40): 68809-68824, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978158

ABSTRACT

Single nucleotide polymorphisms (SNPs) in three microRNAs (miRNAs), rs2910164 in miR-146a, rs11614913 in miR-196a2, and rs3746444 in miR-499, have been associated with breast cancer (BC) susceptibility, but the evidence is conflicting. To obtain a more robust assessment of the association between these miRNA variants and BC risk, we carried out a meta-analysis through systematic literature retrieval from the PubMed and Embase databases. A total of 9 case-control studies on rs2910164, 12 on rs11614913, and 7 on rs3746444 were included. Pooled odds ratios and 95% confidence intervals were used to evaluate associations with BC risk. Overall analysis showed that rs2910164 was not associated with BC susceptibility in any genetic model, whereas rs11614913 was associated with a decreased risk in both the allelic contrast and recessive models, and rs3746444 imparted an increased risk in all genetic models. Stratified analyses showed that rs11614913 may decrease the risk of BC in the heterozygote model in Asians, and in all genetic models, except the heterozygote model, when the sample size is ≥ 500. Subgroup analysis indicated that rs3746444 was associated with increased risk of BC in Asians, but not Caucasians, at all sample sizes. This meta-analysis suggests that rs11614913 in miR-196a2 may decrease the risk of BC, while rs3746444 in miR-499 may increase it, especially in Asians when the sample size is large. We propose that rs11614913(C > T) and rs3746444 (A > G) may be useful biomarkers predictive of BC risk.

2.
Cancer Biol Med ; 13(1): 142-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27144069

ABSTRACT

OBJECTIVE: Survival and treatment of patients with microinvasive breast cancer (MIBC) remain controversial. In this paper, we evaluated whether adjuvant chemotherapy is necessary for patients with MIBC to identify risk factors influencing its prognosis and decide the indication for adjuvant chemotherapy. METHODS: In this retrospective study, 108 patients with MIBC were recruited according to seventh edition of the staging manual of the American Joint Committee on Cancer (AJCC). The subjects were divided into chemotherapy and non-chemotherapy groups. We compared the 5-year disease-free survival (DFS) and overall survival (OS) rates between groups. Furthermore, we analyzed the factors related to prognosis for patients with MIBC using univariate and multivariate analyses. We also evaluated the impact of adjuvant chemotherapy on the prognostic factors by subgroup analysis after median follow-up time of 33 months (13-104 months). RESULTS: The 5-year DFS and OS rates for the chemotherapy group were 93.7% and 97.5%, whereas those for the non-chemotherapy group were 89.7% and 100%. RESULTS indicate that 5-year DFS was superior, but OS was inferior, in the former group compared with the latter group. However, no statistical significance was observed in the 5-year DFS (P=0.223) or OS (P=0.530) rate of the two groups. Most relevant poor-prognostic factors were Ki-67 overexpression and negative hormonal receptors. Cumulative survival was 98.2% vs. 86.5% between low Ki-67 (≤20%) and high Ki-67 (>20%). The hazard ratio of patients with high Ki-67 was 16.585 [95% confidence interval (CI), 1.969-139.724; P=0.010]. Meanwhile, ER(-)/PR(-) patients with MIBC had cumulative survival of 79.3% compared with 97.5% for ER(+) or PR(+) patients with MIBC. The hazard ratio for ER(-)/PR(-) patients with MIBC was 19.149 (95% CI, 3.702-99.057; P<0.001). Subgroup analysis showed that chemotherapy could improve the outcomes of ER(-)/PR(-) patients (P=0.014), but not those who overexpress Ki-67 (P=0.105). CONCLUSIONS: Patients with MIBC who overexpress Ki-67 and with negative hormonal receptors have relatively substantial risk of relapse within the first five years after surgery. However, adjuvant chemotherapy can only improve the outcomes of ER(-)/PR(-) patients, but not those who overexpress Ki-67. Further studies with prolonged follow-up of large cohorts are recommended to assess the prognostic significance and treatment of this lesion.

3.
Cancer Biol Med ; 12(2): 126-39, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26175928

ABSTRACT

OBJECTIVE: A meta-analysis was performed to augment the insufficient data on the impact of mutative EGFR downstream phosphatidylinositol-3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways on the clinical efficiency of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) treatment of non-small cell lung cancer (NSCLC) patients. METHODS: Network databases were explored in April, 2015. Papers that investigated the clinical outcomes of NSCLC patients treated with EGFR-TKIs according to the status of K-ras and/or PIK3CA gene mutation were included. A quantitative meta-analysis was conducted using standard statistical methods. Odds ratios (ORs) for objective response rate (ORR) and hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS) were calculated. RESULTS: Mutation in K-ras significantly predicted poor ORR [OR =0.22; 95% confidence interval (CI), 0.13-0.35], shorter PFS (HR =1.56; 95% CI, 1.27-1.92), and shorter OS (HR =1.59; 95% CI, 1.33-1.91) in NSCLC patients treated with EGFR-TKIs. Mutant PIK3CA significantly predicted shorter OS (HR =1.83; 95% CI, 1.05-3.20), showed poor ORR (OR =0.70; 95% CI, 0.22-2.18), and shorter PFS (HR =1.79; 95% CI, 0.91-3.53) in NSCLC patients treated with EGFR-TKIs. CONCLUSION: K-ras mutation adversely affected the clinical response and survival of NSCLC patients treated with EGFR-TKIs. PIK3CA mutation showed similar trends. In addition to EGFR, adding K-ras and PIK3CA as routine gene biomarkers in clinical genetic analysis is valuable to optimize the effectiveness of EGFR-TKI regimens and identify optimal patients who will benefit from EGFR-TKI treatment.

4.
Curr Cancer Drug Targets ; 13(3): 278-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23369095

ABSTRACT

Dendritic cells (DCs) have traditionally been viewed as constituting an 'information management' system that functions solely to integrate a diverse array of incoming signals, in order to induce immune reactivity. In recent years, however, there has been a shift towards viewing these cells as key regulators in the orchestration of immunological tolerance, with increasing recognition that they are capable of suppressing T-cell responses depending on signalling processes and localised biochemical conditions. Indoleamine 2,3-dioxygenase (IDO) competent (IDO⁺) DCs are a subset of human DCs which are programmed to a tolerogenic state and play a vital role in establishing and maintaining a tumour-suppressing milieu. The expression of IDO in these DCs represents a key mechanism responsible for inducing the tolerogenic state. However, the mechanisms by which IDO becomes dysregulated in this subset of DCs have not yet been described. In this review, the function of IDO⁺ DCs within the cancer-tolerogenic milieu, as well as the signals responsible for expression of IDO in this subset, will be discussed.


Subject(s)
Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CTLA-4 Antigen/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Immune Tolerance/drug effects , Immunologic Surveillance/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , NF-kappa B/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/prevention & control , Protein Isoforms/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/drug effects
5.
Eur J Cell Biol ; 92(2): 54-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23279926

ABSTRACT

MicroRNAs (MiRNAs) are small non-coding RNAs that regulate their target genes expression at the post-transcriptional level. As accumulating properties of miR-205 have been identified, complex roles of miR-205 in tumor initiation and progression are emerging. MiR-205 acts either as a tumor suppressor through inhibiting proliferation and invasion, or as an oncogene through facilitating tumor initiation and proliferation, depending on the specific tumor context and target genes. In this review, we focus on the properties of miR-205 in cancers to shed light on better management of various fatal malignancies. Moreover, we discuss epigenetics that may account for the fluctuation of miR-205 expression. In addition, we sketch a network of miR-205 and its targets to further elucidate the mechanisms through which miR-205 exerts its multiple functions.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasms/genetics , RNA, Neoplasm/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Epithelium/metabolism , Epithelium/pathology , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/diagnosis , Neoplasms/metabolism , Prognosis , RNA, Neoplasm/metabolism
6.
J Innate Immun ; 5(3): 209-18, 2013.
Article in English | MEDLINE | ID: mdl-23207147

ABSTRACT

Interferon (IFN)-λs are a new addition to the old IFN family and share many similarities, such as antiviral and antiproliferative characteristics, with type I IFNs. IFN-λs also exhibit unique characteristics in immunomodulation. Accumulating studies have indicated the interactions between IFN-λs and immune cells, which lead to the regulation of the latter. IFN-λs can influence dendritic cells (DCs) and their product, IFN-λs-DCs, can then regulate the function of T cells. On the other hand, IFN-λs can also directly affect T cells through inhibition of the T helper 2 cell (Th2) responses. IFN-λs have varying immunomodulatory functions under different physiological conditions or in different organs and can inhibit tumor growth via regulation of the immune system. Diseases associated with IFN-λs include asthma, allergy, and systemic lupus erythematosus. In this review, we summarize the current knowledge of the biology of IFN-λs and their immunomodulatory function in relevant human diseases.


Subject(s)
Asthma/immunology , Immunologic Factors/immunology , Interleukins/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Asthma/drug therapy , Dendritic Cells/immunology , Humans , Immunologic Factors/therapeutic use , Interferons , Interleukins/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Th2 Cells/immunology
7.
J Cancer Res Clin Oncol ; 138(12): 2069-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22821179

ABSTRACT

PURPOSE: Non-small-cell lung cancer (NSCLC) cells with somatic mutations in epidermal growth factor receptors (EGFR) are initially susceptible to tyrosine kinase inhibitor (TKI); however, eventually resistance to TKI is developed in these cells, which leads to the failure of treatment. The most common mechanism of this acquired drug resistance is development of a secondary T790M mutation in EGFR. In this study, we investigated the effects of the combination of Erlotinib and Cetuximab on T790M and L858R mutation lung cancer cells lines (H1975), in the primary NSCLC cells with the T790M mutation and TKI-resistant EGFR mutations human tumor xenograft model (H1975). METHODS: The effects of these two agents on cell proliferation, apoptosis, and EGFR-dependent signaling were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, annexin V staining, and Western blotting. Sensitivity of EGFR inhibitors was detected in the primary tumor cell suspension and human tumor xenograft model (H1975). RESULTS: Compared with single-agent treatment, the combination of Cetuximab and Erlotinib increased apoptosis of EGFR TKI-resistant NSCLC cells (H1975), resulting in more pronounced growth inhibition on cell proliferation and significant inhibition of EGFR-dependent signaling. CONCLUSIONS: These data suggest that treatment with a combination of Erlotinib and Cetuximab overcomes T790M-mediated drug resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Erlotinib Hydrochloride , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mutation/drug effects , Quinazolines/administration & dosage , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays
8.
Biochem Biophys Res Commun ; 420(4): 699-703, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22465008

ABSTRACT

Tumor stem cells (TSCs) are considered as the "seeds" in tumor development, metastasis and recurrence. Despite the various immunosurveillance mechanisms in the host, TSCs may possess the phenotypic and functional properties to evade host immunosurveillance and immune-mediated rejection in immunologically intact individuals. The mechanisms of TSC recognition and their consequent destruction are actively disturbed by various processes, including altered immunogenicity of TSCs, production of TSC-derived regulatory molecules, and interaction of TSCs with tumor-infiltrating immune cells. In addition to these TSC-mediated mechanisms, the diverse mesenchymal cells and cytokines in the tumor microenvironment are contribute to TSC immune escape. Recent mechanistic studies provide a more comprehensive understanding of TSCs in the biology, prevention, and therapy of solid tumors. This review will focus on the latest findings for mechanisms underlying TSCs' escape from the attack of immune system.


Subject(s)
Immunologic Surveillance , Neoplastic Stem Cells/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Humans , Immune System/immunology
9.
Int J Cancer ; 130(9): 1971-80, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22025331

ABSTRACT

SOCS1, a prototype molecule of the SOCS family, was initially defined as a suppressor of cytokine signaling. The molecular mechanisms of SOCS1-mediated functions have been subsequently identified by studies using gene knockout mice and gene silencing technology. As part of a negative feedback regulation, SOCS1 downregulates cytokine signaling through direct inhibition of the JAK tyrosine kinase and the signaling cascade of activated cytokine receptors, thereby attenuating cytokine-initiated signal transduction. Moreover, other studies have demonstrated that SOCS1 also downregulates TLR signaling through direct and indirect mechanisms. Both cytokine receptor and TLR signaling pathways mediate important functions in survival, maturation and differentiation of various types of cells and in the regulation of immune function. Abnormal expression of SOCS1 in tumor cells has been detected in various human cancers, where it is associated with dysregulation of cytokine receptor and TLR signaling to promote cell transformation. Recent studies on the function of SOCS1 in tumor cells have revealed its novel role in carcinogenesis. In this review, we will focus on the mechanism of action of SOCS1 in both tumor cells and antigen-presenting cells in the tumor microenvironment. The potential of using SOCS1 as a diagnostic marker and therapeutic target in tumor diagnosis, prognosis and treatment is discussed.


Subject(s)
Neoplasms/metabolism , Receptors, Cytokine/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Toll-Like Receptors/metabolism , Animals , Biomarkers, Tumor/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mice , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/genetics , Prognosis , Receptors, Cytokine/genetics , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/genetics , Toll-Like Receptors/genetics
10.
Zhonghua Zhong Liu Za Zhi ; 33(7): 513-6, 2011 Jul.
Article in Chinese | MEDLINE | ID: mdl-22093628

ABSTRACT

OBJECTIVE: To investigate the expression of indoleamine 2, 3-dioxygenase (IDO) in breast cancer and its correlation with clinicopathologic factors and prognosis. METHODS: The expression of IDO, CD31, CD105 proteins in 40 specimens of breast cancer were assessed by immunohistochemistry. RESULTS: The overexpression rate of IDO in breast cancer was 67.5% (27/40), and expression of IDO was closely associated with clinical stage and lymph nodes metastasis. The disease-free survival rate in patients with IDO overexpression was not significantly lower than that in patients with negative or low expression of IDO (P > 0.05). Moreover, the expression of IDO was positively correlated with CD105-labeled microvessel density (r = 0.659, P < 0.05). CONCLUSIONS: Expression of IDO is associated with clinical stage and lymph nodes metastasis, and microvessel densitty. IDO expression may promote the growth and metastasis of breast cancer, probably via the increased agiogenesis. A larger sample study is needed to verify whether the prognosis of beast cancer is significantly correlated with IDO expression.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Microvessels , Adenocarcinoma/enzymology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adult , Aged , Antigens, CD/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Medullary/enzymology , Carcinoma, Medullary/immunology , Carcinoma, Medullary/pathology , Disease-Free Survival , Endoglin , Female , Follow-Up Studies , Humans , Immunohistochemistry , Lymphatic Metastasis , Microvessels/enzymology , Microvessels/immunology , Middle Aged , Neoplasm Staging , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Cell Surface/metabolism , Survival Rate
11.
Cancer Biother Radiopharm ; 26(4): 503-10, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21812652

ABSTRACT

Traditional immunotherapy for patients with refractory metastatic renal cell carcinoma (RCC) is limited because the tumors themselves induce immunosuppression. The aim of this article was to evaluate the clinical efficacy of the infusion of a high dose of interleukin (IL)-2-activated allogeneic haploidentical peripheral blood stem cells (haplo-PBSCs) in patients with advanced intractable RCC. Ten advanced RCC patients and their haploidentical relatives, who were haplo-PBSC donors, were enrolled in this study. All patients accepted one cycle of activated haplo-PBSCs. The clinical and immunologic responses were evaluated. A range from 2.3 to 5.5×10(10) of activated haplo-PBSCs were harvested after exposure to recombinant human IL-2 (rhIL-2), along with a significant increase in the proportion of natural killer cells and activated lymphocytes (CD69+ and CD25+). Enhanced cytotoxicity of haplo-PBSCs for RCC was also observed. After treatment, 2 (2/10) cases of partial remission, 6 (6/10) cases of stable disease, and 2 (2/10) cases of progressive disease were identified in these 10 patients. The median progression-free survival of the 10 patients was 5.5 months (3-14 months). The adoptive transfusion of IL-2-activated haplo-PBSCs can induce sustained antitumor effects for advanced intractable RCC patients who have had no response to conventional immunotherapy.


Subject(s)
Carcinoma, Renal Cell/therapy , HLA Antigens/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Immunotherapy, Adoptive/methods , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Aged , Aged, 80 and over , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Female , Haplotypes , Humans , Interleukin-2/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Treatment Outcome
12.
Chin Med J (Engl) ; 123(16): 2215-20, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20819668

ABSTRACT

BACKGROUND: S100A8 and S100A9 are two members of the S100 protein family characterized by the presence of two Ca2+-binding sites of the EF-hand type. Previous studies suggested that the whole S100 family displays significant functions in tumor growth, progression and invasion. This study aimed to determine the expression of the two indices of the family, S100A8 and S100A9, in lung cancer tissues and normal lung tissues and its correlation with clinical features. METHODS: A total of 60 cases with a variety of clinical data that were diagnosed with different histological subtypes of lung cancer were investigated. Semi-quantitative reverse transcriptase-PCR (Sq-Rt-PCR) and immunohistochemical staining of cancer, adjacent and peripheral lung tissues were executed to distinguish the expression patterns of S100A8 and S100A9 and to further clarify their correlation with clinical features. RESULTS: Immunohistochemical staining of both proteins showed a significant up-regulation in lung cancer tissue (S100A8, S100A9, P<0.0001), and PCR revealed that the levels of S100A8 and S100A9 expression were significantly higher in lung cancer tissues (S100A8 P=0.002/0.004; S100A9 P=0.022/0.026). The higher expression was found to be correlated with the clinical characteristics of adenocarcinoma, inflammation and stage IV lesion. CONCLUSIONS: S100A8, S100A9 up-regulation was found in the lung adenocarcinoma and end stage lung cancer tissue, the correlation of which with their higher expression in inflammatory lung tissues may indicate the collaborative effect of inflammation on the progression of cancer.


Subject(s)
Adenocarcinoma/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism , Inflammation/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aged , Calgranulin A/genetics , Calgranulin B/genetics , Female , Humans , Immunohistochemistry , Inflammation/genetics , Inflammation/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
13.
Zhonghua Yi Xue Za Zhi ; 89(6): 423-6, 2009 Feb 17.
Article in Chinese | MEDLINE | ID: mdl-19567126

ABSTRACT

OBJECTIVE: To further identify the breast adenocarcinoma cell line T47D-tk stably expressing the suicide gene HSV1-tk, observe its growing characteristics, and establish an animal model of implanted breast adenocarcinoma. METHODS: Retroviral vector of HSV1-tk gene and breast carcinoma cell line T47D-tk stably expressing the HSV1-tk gene were established. Breast carcinoma cells of the lines T47D and T47D-tk were cultured, and observed by inverted microscope. Growth curve was drawn. Genomic DNA of T47D-tk cells was extracted, and amplified by PCR with HSV1-tk and HSV1-tk P2 as primers. Agarose gel electrophoresis was used to identify the HSV1tk gene. Suspensions of T47D and T47D-tk cells were inoculated subcutaneously at bilateral roots of foreleg of female BALB/c nude mice respectively. The growth of tumor was observed every day. RESULTS: PCR showed 1131 bp fragment in the T47D-tk genome, but not in the T47D genome. The numbers of growing T47D cells on days 3, and 7 were (10.00 +/- 1.30) x 10(3) and (19.25 +/- 0.66) x 10(3) respectively, not significantly different from those of the T47D-tk cells [(10.25 +/- 0.90) x 10(3) and (19.00 +/- 1.80) x 10(3) respectively, both P > 0.05]. The time needed for tumor formation after the inoculation of T47D cells was (9.67 +/- 0.33) d, not significantly different from that of the T47D-tk cells [(9.83 +/- 0.48) d, P > 0.05]. The tumor size 19 days after inoculation of the T47D cells was (72.17 +/- 25.88) mm(3), not significantly different from that of the T47D-tk cells [(70.66 +/- 22.16) mm(3), P > 0.05]. CONCLUSION: The T47D-tk cells have integrated the HSV1-tk gene without changing its growing characteristics. An animal model of implanted breast cancer has been successfully established. The T47D and T47D-tk subcutaneous xenografts offer the foundation for further study of gene imaging and gene therapy.


Subject(s)
Breast Neoplasms/genetics , Genes, Transgenic, Suicide/genetics , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Therapy , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Transfection
14.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 17(1): 164-9, 2009 Feb.
Article in Chinese | MEDLINE | ID: mdl-19236771

ABSTRACT

This study was aimed to study the potential effects of alloreactive NK cells (allo-NKs) in therapy of relapsed lung cancer after haploidentical hematopoietic stem cell transplantation using donor lymphocyte infusion (DLI). The F1 donors derived-NK cells were purified with MACS magnetic separation system, in which the proportion of the alloreactive Ly49A(+) cells was detected by flowcytometry and alloreactivity was measured by LDH method. The relapse model of lung cancer after haploidentical-HSCT was established. The distribution kinetic of infused donor lymphocytes in vivo was analyzed. The inhibition of relapse tumor, infiltration of lymphocytes in situ and fluctuation of 22 kinds of cytokines in serum after DLI were compared among different groups. The results showed that the infused donor cells of allo-NK group were accumulated mostly in lung, spleen and kidney for more than 48 hours with considerable higher levels according to the distribution kinetic curve. The sizes of relapse tumors between chemotherapy + PBS group and chemotherapy + DLI group showed no difference. However, the relapsed tumors in allo-NK + DLI group were significantly smaller than that in chemotherapy + DLI group or allo-NK + PBS group, in which increased infiltration of lymphocytes were defined in situ. The levels of cytokines such as MCP-1, IL-17, IL-12 and MCP-5 in serum of allo-NK + DLI group ascended compared with control group, though the level of IL-10 declined simultaneously. It is concluded that allo-NKs prolong the survival time of infused donor lymphocytes in vivo, promote the secretion of inflammatory cytokines and Th1-type of cytokines, and further improve the antitumor effects of DLI against relapse after transplantation.


Subject(s)
Killer Cells, Natural/cytology , Lung Neoplasms/therapy , Lymphocyte Transfusion/methods , Neoplasm Recurrence, Local/therapy , Transplantation Conditioning/methods , Animals , Cytokines/blood , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
15.
Zhonghua Nei Ke Za Zhi ; 48(10): 857-61, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-20079231

ABSTRACT

OBJECTIVE: To study the effect of feto-maternal microchimerism in the treatment of activated human leukocyte antigen (HLA) haploidentical mobilized peripheral blood cells against solid tumors. METHODS: Genomic DNA samples of 25 pairs of HLA haploidentical donors and recipients were extracted. The donor-derived HLA-DRB loci were detected with nested PCR-sequence specific primer (SSP) typing. The mixed lymphocyte proliferation action between the patients and respective donors, the engraftment of donor's cells and the serum levels of Th1/Th2 type of cytokines were measured with MTT, FISH and ELISA method respectively. The survival time of patients with or without feto-maternal microchimerism were compared as well. RESULTS: Using nested PCR-SSP typing, the positive rates of feto-maternal microchimerism in the 25 pairs of HLA haploidentical donors and recipients were 40% in the maternal/children pairs and 0 in the paternal/children pairs. The chimerism positive patients showed less proliferation activity when cocultured with respective donors as compared with unrelated ones (P = 0.03). Only one chimerism positive patient experienced the engraft of donor's cell 3 months after treatment as the donor derived XX chromosome was identified with FISH. When the data of chimerism positive patients were deleted, the serum levels of IFNgamma 1 month after treatment dropped dramatically from 171.4 (26.3 approximately 258.4) ng/L to 29.4 (1.2 approximately 39.9) ng/L. The survival time in chimerism positive patients of the maternal/children pairs was significantly longer than that in chimerism negative patients, which was (31.2 +/- 4.3) months and (11.1 +/- 3.3) months, respectively (P = 0.036). CONCLUSION: Feto-maternal microchimerism might induce anergy in the HLA haploidentical donors, favor the engraftment of donor's progenitors and maintenance of positive microenvironment and prolong the survival time.


Subject(s)
Chimerism , HLA Antigens/genetics , Neoplasms/genetics , Peripheral Blood Stem Cell Transplantation , Transplantation Conditioning/methods , Adult , Aged , Female , Graft Rejection/prevention & control , HLA Antigens/immunology , Haploidy , Histocompatibility Testing , Humans , Male , Middle Aged , Neoplasms/immunology , Neoplasms/therapy , Transplantation, Homologous
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 24(11): 1040-3, 2008 Nov.
Article in Chinese | MEDLINE | ID: mdl-18992186

ABSTRACT

AIM: To construct eukaryotic expression vector and express human interleukin 18 (hIL-18) in Pichia pastoris. METHODS: The gene encoding of hIL-18 was amplification by PCR. The recombinant pPICZaC/hIL-18 was transformed into the Pichia pastoris X-33 strain via electroporation. The high level expression was selected and assayed by the methods of PCR, SDS-PAGE and Western blot. The rhIL-18 was purified by the methods of hydrophobic chromatography and anion exchange chromatography. The bioactivity of it was initially assayed. RESULTS: The rhIL-18 was secreted into the supernatant and the concentration reached to 202 mg/L. The rhIL-18 was further identified by Western blot with specific antibody binding activity. The purity of the rhIl-18 reached about 95%. And rhIL-18 can synergistically induce PBMC to produce IFN-gamma with IL-2. CONCLUSION: A rhIL-18 is successfully constructed and expressed in Pichia pastoris. And this contributes to further study of its function and activity.


Subject(s)
Interleukin-18/metabolism , Pichia/metabolism , Recombinant Proteins/metabolism , Blotting, Western , Cells, Cultured , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Humans , Interferon-gamma/metabolism , Interleukin-18/genetics , Interleukin-18/isolation & purification , Interleukin-18/pharmacology , Interleukin-2/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Pichia/genetics , Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
17.
Cell Immunol ; 253(1-2): 59-65, 2008.
Article in English | MEDLINE | ID: mdl-18522858

ABSTRACT

Objective. To investigate the phenotype changes and proliferation activities of cytokine-induced killer cells (CIKs) and lymphokine-activated killer cells (LAKs) from healthy donor, and the cytotoxicities of CIKs and LAKs to human in vitro glioma cell lines U251 and U87. Therapy of CIK intratumoral injection was evaluated in nude mouse models. Methods. CIK cells were induced from peripheral blood mononuclear cells (PBMC) of healthy donors with multiple cytokines. Phenotype analysis of CIKs and LAKs was performed with flow cytometer (FCM). The specific cytotoxicities of CIKs and LAKs against cell line U251 and U87 were determined by LDH method. After intracerebral injection of CIKs, the distribution of CIKs and the inflammatory reaction of their surrounding brain tissue were observed through continuous pathological sections. In vivo anti-tumor activity of CIKs was evaluated in athymic nude mice with intracerebral xenotransplanted U251 glioma by MRI. Results. Amount of CIKs was increased (49.83+/-2.04) times and double positive cells, CD3(+)/CD56(+) cells, were increased from (3.36+/-1.85%) to (44.07+/-14.14%) with elevated absolute amount over 1000 times after 2 week culture. In vitro experiments demonstrated that compared with LAK, CIKs possessed more obvious cytotoxic activity to U251 and U87. In vivo experiments showed that there was no severe inflammatory reaction in brain tissue. CIKs can markedly inhibit intracranial xenotransplanted glioma growth by intracranial injection (P<0.01). Conclusion. CIKs are a kind of highly effective immune cells which have a strong suppressive effect on growth for in vitro and in vivo glioma. Local injection of CIKs does not produce severe damage to normal brain tissue and is likely to be used in clinical adoptive immunotherapy of intracerebral glioma.


Subject(s)
Brain Neoplasms , Cytokines/immunology , Glioma , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Neoplasm Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Proliferation , Glioma/immunology , Glioma/pathology , Glioma/therapy , Humans , Killer Cells, Natural/cytology , Mice , Mice, Nude , Phenotype , Tumor Cells, Cultured
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 15(5): 1013-8, 2007 Oct.
Article in Chinese | MEDLINE | ID: mdl-17956681

ABSTRACT

This study was aimed to investigate the feasibility of low dose of fludarabine, cyclophosphamide combined with donor derived alloreactive NK cells as a new nonmyeloablative conditioning regimen in the haploidentical hematopoietic stem cell transplantation (haploidentical HSCT). F1 derived-NK cells were enriched with MACS magnetic separation system, in which the proportions of the Ly49C+ and Ly49A+ cells were detected by flow cytometry and the alloreactivity was measured by LDH method. The haploidentical HSCT models were constructed, and the myeloablativity in vivo, donor engraftment and the intensity of GVHD were compared between different myeloablative and nonmyeloablative conditioning regimens, including 9 Gy TBI, 6.5 Gy TBI, flu + cy, and flu + cy + allo-NK. The results showed that the flu + cy + allo-NK conditioning was nonmyeloablative, but the rate of donor chimerism after haploidentical HSCT was significantly higher as compared with other nonmyeloablative methods, which were (28.70 +/- 5.90)% in bone marrow and (46.40 +/- 5.00)% in spleen at day 21 post-transplantation. When compared with the flu + cy conditioning, the intensity of GVHD was slight in the flu + cy + allo-NK group, in which only a half of C57BL/6 recipients experienced weight loss, and no distinct pathological damages observed in the liver, intestine, kidney and skin samples. It is concluded donor derived-alloreactive NK cells can facilitate engraftment of the haploidentical hematopoietic stem cells and mitigate GVHD. The flu + cy + allo-NK conditioning provides a new method for those elder patients with high-risk solid tumor undergoing haploidentical-HSCT.


Subject(s)
Cyclophosphamide/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Killer Cells, Natural/transplantation , Transplantation Conditioning/methods , Vidarabine/analogs & derivatives , Animals , Female , Graft vs Host Disease/prevention & control , Haplotypes , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Transplantation Chimera , Vidarabine/administration & dosage , Whole-Body Irradiation
19.
Cancer Biother Radiopharm ; 22(2): 223-34, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17600469

ABSTRACT

OBJECTIVE: The traditional immunotherapy for patients with refractory metastatic solid tumors is limited because tumors induce immunosuppression. New treatment is, therefore, needed. The aim of this study was to evaluate the clinical efficacy of infusion of high-dose interleukin (IL)-2-activated allogeneic haploidentical peripheral blood stem cells (haplo-PBSCTs) on patients with an advanced stage of refractory solid tumors. METHODS: This study involved 11 patients with refractory metastatic tumors and haploidentical relatives as donors for haplo-PBSCs. The therapeutic outcome of the IL-2-activated haplo-PBSC infusion and patients' cytokine levels were evaluated. The cytotoxicity of IL-2-activated haplo-PBSCs for tumor cells was determined using in vitro cytotoxicity assays. RESULTS: A range from 2.5 to 5.6 x 10(10) of activated haplo-PBSCs were harvested after exposure to rhIL-2, along with a significant increase in the proportion of natural killer (NK) cells and activated lymphocytes (CD69+ and CD25+), and enhanced cytotoxicity of haplo-PBSCs for several tumor cell lines. Following treatment, 1 (1/11) patient achieved a partial response (PR), 1 (1/11) achieved a mild response (MR), 6 (6/11) achieved stable disease (SD), and 3 (3/11) achieved progressive disease (PD). For all of the 11 patients, the median progression-free survival (PFS) was 5 months (3-14 months). We also observed the phenomenon of Th2 shifted to Th1, which played a crucial role in cancer immunotherapy. CONCLUSIONS: The adoptive transfusion of IL-2-activated haplo-PBSCs has potent antitumor effects both in vitro and in vivo. This finding suggests that IL-2-activated haplo-PBSCs may serve as an alternative therapy for advanced-stage solid tumors, especially for those patients who are refractory or ineligible for chemo- or radiotherapy.


Subject(s)
HLA Antigens/genetics , HLA Antigens/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Interleukin-2/pharmacology , Neoplasms/pathology , Neoplasms/therapy , Adult , Aged , Blood Donors , Cell Differentiation , Female , Haplotypes/genetics , Humans , Immunotherapy , Male , Middle Aged , Neoplasm Metastasis/pathology , Phenotype , Th1 Cells/cytology , Th2 Cells/cytology , Tomography, X-Ray Computed , Treatment Outcome
20.
J Clin Immunol ; 27(3): 317-26, 2007 May.
Article in English | MEDLINE | ID: mdl-17468835

ABSTRACT

: CD(4) (+)CD(25) (+) regulatory T cells (Tregs) have been shown to inhibit cytotoxic lymphocytes-mediated immune responses. Cytokine-induced killer (CIK) cells exert high impact on adoptive immunotherapeutic approaches. Therefore, the purpose of this report was to determine the effect of Tregs on CIK cell growth and CIK-induced cytotoxicity for inhibition of tumor growth in vivo as well as in vitro. After depletion of CD(4) (+)CD(25) (+) cells before culture, the proliferation and cytotoxicity of CIK cells, which indicated in bromodeoxyuridine (BrdU) and lactic dehydrogenase (LDH) assays, were significantly increased. Depletion of CD(4) (+)CD(25) (+) cells preculture also enhanced the suppression effect on the lung cancer cells inoculated in experimental animals. Blockage of glucocorticoid-induced tumor necrosis factor receptor (GITR) and transforming growth factor beta1 (TGF-beta1) by antibodies partially abrogated the suppressive effect of CD(4) (+)CD(25) (+) cells on CIK. These results indicated that Tregs could inhibit the antitumor activity of CIK cells. The molecules TGF-beta and GITR may contribute to the suppressive function of CD(4) (+)CD(25) (+) cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Animals , Antibodies/immunology , Cell Proliferation , Humans , Killer Cells, Natural/cytology , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Phenotype , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...