Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncoimmunology ; 6(7): e1328335, 2017.
Article in English | MEDLINE | ID: mdl-28811965

ABSTRACT

Our previous phase I/IIA study showed that autologous dendritic cells (DCs) pulsed with tumor-associated antigens are well tolerated in patients with hepatocellular carcinoma (HCC). In this randomized, multicenter, open-label, phase II trial, we investigated the efficacy and safety of this DC-based adjuvant immunotherapy with 156 patients, who treated for HCC with no evidence of residual tumor after standard treatment modalities. Patients were randomly assigned to immunotherapy (n = 77; injection of 3 × 107 DC cells, six times over 14 weeks) or control (n = 79; no treatment). The primary end point was recurrence-free survival (RFS), and the secondary endpoints were immune response and safety. The RFS between the immunotherapy and control groups was not significantly different (hazard ratio [HR], 0.97; 95% confidence interval [CI], 0.60-1.56; p = 0.90). However, post-hoc subgroup analyses revealed that DC immunotherapy significantly reduced the risk of tumor recurrence of non-radiofrequency ablation (non-RFA) group patients (n = 83, HR, 0.49; 95% CI, 0.26-0.94; p = 0.03), whereas unexpectedly increased the risk of recurrence in RFA group (n = 61, p = 0.01). Tumor-specific immune responses were significantly enhanced (both p < 0.01) in the immunotherapy group. Baseline serum interleukin (IL)-15 was statistically correlated with RFS prolongation (HR, 0.16; 95% CI, 0.03-1.58; p = 0.001) within the immunotherapy groups. Overall adverse events were more frequent in the immunotherapy group (p < 0.001) but were mainly mild to moderate in severity. In conclusion, adjuvant immunotherapy with DC vaccine reduces the risk of tumor recurrence in HCC patients who underwent standard treatment modalities other than RFA. Baseline IL-15 might be a candidate biomarker for DC-based HCC immunotherapy.

2.
Br J Cancer ; 113(12): 1666-76, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26657650

ABSTRACT

BACKGROUND: To date, no adjuvant treatment has been shown to have a clear benefit in patients with hepatocellular carcinoma (HCC). In this prospective phase I/IIa study, we evaluated the safety and efficacy of adjuvant dendritic cell (DC) therapy in HCC patients who received primary treatment for HCC. METHODS: Twelve HCC patients who had no viable tumour after primary treatments were included. Dendritic cell vaccines pulsed with cytoplasmic transduction peptide-attached alpha-fetoprotein, glypican-3 and melanoma-associated antigen 1 recombinant fusion proteins were injected subcutaneously near to inguinal lymph nodes. Adverse effects, time to progression (TTP), and associated immune responses were evaluated after DC vaccination. RESULTS: Nine of 12 patients had no tumour recurrence up to 24 weeks after DC vaccination. Among a total of 144 adverse events, 129 events (89.6%) were regarded as adverse drug reactions, all of which were grade 1 or 2. The majority of patients showed enhanced anti-tumour immune responses after DC vaccination. Recurrence-free patients exhibited relatively stronger anti-tumour immune responses than patients who developed recurrence after DC vaccination, as evidenced by lymphocyte proliferation and IFN-γ ELISPOT assays. The median time of TTP was 36.6 months in the DC-vaccination group and 11.8 months in the control group (hazard ratio, 0.41; 95% confidence interval, 0.18-0.95; P=0.0031 by log-rank test). CONCLUSIONS: Adjuvant DC vaccine for HCC was safe and well tolerated in phase I/IIa study, and preliminary efficacy data are encouraging to warrant further clinical study in patients with HCC after primary treatments.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma, Hepatocellular/therapy , Cell Transplantation , Dendritic Cells/immunology , Immunotherapy , Liver Neoplasms/therapy , Aged , Female , Humans , Male , Middle Aged , Treatment Outcome
3.
J Surg Oncol ; 93(6): 504-10, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16615154

ABSTRACT

BACKGROUND AND OBJECTIVES: Although more than five variant forms of estrogen receptor-beta (ERbeta) have been identified, their role has not been identified. This study was carried out to investigate the changes of ERbeta variants in breast cancer development. METHODS: Using reverse transcription polymerase chain reaction (RT-PCR) and triple primer PCR (TP-PCR), the expression levels of ERbeta variants mRNA were measured in 66 paired normal and cancer tissues. The relative expression level of ERbeta variants were compared between normal and cancer tissues, and also compared according to various clinicopathological parameters. RESULTS: Among ERbeta variants, ERbeta2 and ERbeta5 consist of the major proportion of ERbeta expression both in normal and cancer tissues. The ERbeta and ERbeta2 expression levels decreased significantly in the cancers compared with corresponding normal tissues, particularly in ERalpha-expressing cancers. However, ERbeta5 expression level increased significantly in the cancers, especially in those of postmenopausal patients. The relative increase of ERbeta5 expression in cancer tissues was associated with favorable differentiation. CONCLUSIONS: Decrease of ERbeta2 is thought to be the key reason for the decrease in ERbeta expression in cancer tissues, and it is particularly associated with the development of ERalpha-expressing breast cancer.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Estrogen Receptor beta/metabolism , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/physiology , Female , Humans , Polymerase Chain Reaction , Postmenopause/metabolism , Premenopause/metabolism , RNA Splicing/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction
4.
J Biol Chem ; 280(30): 27783-91, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15917220

ABSTRACT

The POZ domain is a highly conserved protein-protein interaction motif found in many regulatory proteins. Nuclear factor-kappaB (NF-kappaB) plays a key role in the expression of a variety of genes in response to infection, inflammation, and stressful conditions. We found that the POZ domain of FBI-1 (factor that binds to the inducer of short transcripts of human immunodeficiency virus-1) interacted with the Rel homology domain of the p65 subunit of NF-kappaB in both in vivo and in vitro protein-protein interaction assays. FBI-1 enhanced NF-kappaB-mediated transcription of E-selectin genes in HeLa cells upon phorbol 12-myristate 13-acetate stimulation and overcame gene repression by IkappaB alpha or IkappaB beta. In contrast, the POZ domain of FBI-1, which is a dominant-negative form of FBI-1, repressed NF-kappaB-mediated transcription, and the repression was cooperative with IkappaB alpha or IkappaB beta. In contrast, the POZ domain tagged with a nuclear localization sequence polypeptide of FBI-1 enhanced NF-kappaB-responsive gene transcription, suggesting that the molecular interaction between the POZ domain and the Rel homology domain of p65 and the nuclear localization by the nuclear localization sequence are important in the transcription enhancement mediated by FBI-1. Confocal microscopy showed that FBI-1 increased NF-kappaB movement into the nucleus and increased the stability of NF-kappaB in the nucleus, which enhanced NF-kappaB-mediated transcription of the E-selectin gene. FBI-1 also interacted with IkappaB alpha and IkappaB beta.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/physiology , E-Selectin/metabolism , Gene Expression Regulation , NF-kappa B/metabolism , Transcription Factors/physiology , Active Transport, Cell Nucleus , Blotting, Western , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Genes, Dominant , HeLa Cells , Humans , I-kappa B Proteins/metabolism , Immunoblotting , Immunoprecipitation , Leupeptins/pharmacology , Luciferases/metabolism , Microscopy, Confocal , Models, Biological , NF-KappaB Inhibitor alpha , Peptides/chemistry , Plasmids/metabolism , Proline/analogs & derivatives , Proline/pharmacology , Protein Binding , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/chemistry , Tetradecanoylphorbol Acetate/pharmacology , Thiocarbamates/pharmacology , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , Two-Hybrid System Techniques
5.
Breast Cancer Res Treat ; 80(1): 79-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12889601

ABSTRACT

Using messenger RNA (mRNA) in situ hybridization, we investigated estrogen receptor-beta (ERbeta) mRNA levels in normal mammary, benign breast tumor (BBT), breast cancer (BC), and metastatic lymph node tissues to verify the role of ERbeta in BC development and progression. ERbeta expression was significantly decreased in BC and metastatic lymph node tissues compared with normal mammary and BBT tissues (p < 0.01). The intensity and extent of ERbeta mRNA signals were also significantly lower in BC and metastatic lymph node tissues than in the normal mammary and BBT tissues (p < 0.01). An inverse relationship was found between ERbeta mRNA level and both histologic grade (p = 0.091) and progesterone receptor expression (p = 0.052) with marginal significance, but no significant association was noted between ERbeta expression in cancer tissues and the other clinico-pathologic data. The 3-year distant relapse-free survival probability was found to be independent of ERbeta expression. Collectively, ERbeta mRNA decreases in the process of BC development, but seems to be associated with poor differentiation.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Breast Neoplasms/genetics , Disease-Free Survival , Estrogen Receptor beta , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , Lymphatic Metastasis , Prognosis , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Receptors, Estrogen/genetics
6.
Yonsei Med J ; 44(1): 58-64, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12619176

ABSTRACT

A humanized monoclonal antibody against HER2 has been using in a clinical setting and has been shown to possess therapeutic properties. A mimetic peptide against HER2 was also reported to bind to the HER2 receptor with some therapeutic potential. Based on a previous report and the sequence of Herceptin, we designed oligonucleotides of anti-HER2 mimetic peptides, named V2 and V3 peptides, in order to develop a peptide- producing vector system for biologic therapy against HER2- overexpressing cancers. We also adopted the sequence of a previously reported mimetic peptide, V1 (Park BW et al. Nat. Biotechnol, 2000, 18:194-198), as a reference peptide. We examined the effects of the V2 and V3 peptides against the HER2-overexpressing cell lines, SK-BR-3 and T6-17. Transient transfection of the construct expressing V1 and V2 inhibited cell proliferation in HER2-overexpressing cell lines by 20 - 30%, but had no effect on the HER2-negative NIH3T3 cells. The proliferation inhibition shown by V2 was slightly better than that shown by V1. Recombinant peptides V2 and V3 were produced on a large scale in an E. coli system, and the V2 peptide showed anti-HER2-specific tumor cell proliferation inhibition of 10% to 30%. Current results suggest that anti-HER2 mimetic peptides, overexpressed by a constitutive promoter or produced in an E. coli system, could specifically inhibit the proliferation of HER2-expressing cancer cells. Further efforts to augment the biologic specificity and efficacy and to develop new technologies for the purification of the peptide from the E coli system are needed.


Subject(s)
Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Receptor, ErbB-2/chemistry , Technology, Pharmaceutical , Amino Acid Sequence , Animals , Cell Division/drug effects , Cell Line , Mice , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...