Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cancer Immunol Immunother ; 69(5): 825-834, 2020 May.
Article in English | MEDLINE | ID: mdl-32060687

ABSTRACT

Adjuvant cytokine-induced killer (CIK) cell immunotherapy has shown potential in improving the prognosis of hepatocellular carcinoma (HCC) patients after curative resection. However, whether an individual could obtain survival benefit from CIK cell treatment remains unknown. In the present study, we focused on the characteristics of CIK cells and aimed to identify the best predictive biomarker for adjuvant CIK cell treatment in patients with HCC after surgery. This study included 48 patients with HCC treated with postoperative adjuvant CIK cell immunotherapy. The phenotype activity and cytotoxic activity of CIK cells were determined by flow cytometry and xCELLigence™ Real-Time Cell Analysis (RTCA) system, respectively. Correlation analysis revealed that the cytotoxic activity of CIK cells was significantly negative correlated with the percentage of CD3+ CD4+ cell subsets, but significantly positive correlated with CD3-CD56+ and CD3+ CD56+ cell subsets. Survival analysis showed that there were no significant associations between patients' prognosis and the phenotype of CIK cells. By contrast, there was statistically significant improvement in recurrence-free survival (RFS) and overall survival (OS) for patients with high cytotoxic activity of CIK cells as compared with those with low cytotoxic activity of CIK cells. Univariate and multivariate analyses indicated that CIK cell cytotoxicity was an independent prognostic factor for RFS and OS. In conclusion, a high cytotoxic activity of CIK cells can serve as a valuable biomarker for adjuvant CIK cell immunotherapy of HCC patients after surgery.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cytokine-Induced Killer Cells/transplantation , Cytotoxicity, Immunologic , Immunotherapy/methods , Liver Neoplasms/therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Cell Culture Techniques , Cells, Cultured/immunology , Cells, Cultured/transplantation , Combined Modality Therapy/methods , Cytokine-Induced Killer Cells/immunology , Cytotoxicity Tests, Immunologic , Female , Flow Cytometry , Hepatectomy , Humans , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Male , Middle Aged , Postoperative Period , Prognosis , Survival Analysis , Transplantation, Autologous/methods
2.
Mol Carcinog ; 55(1): 64-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25557723

ABSTRACT

Ubiquitination factor E4B (UBE4B) has been speculated to have contradictory functions upon tumorigenesis as an oncogene or tumor suppressor in different types of cancers. We investigated the expression and prognostic role of UBE4B in primary hepatocellular carcinoma (HCC) using cell lines and 149 archived HCC samples. Correlation between the functions of UBE4B in HCC was also explored. We used human HCC cell lines (HepG2, Hep3B, SK-Hep1, Huh7, SMMC-7721, BEL-7402) and a normal hepatocyte cell line (LO2) along with HCC samples from patients who had undergone resection for HCC previously at our hospital. A battery of methods (real-time quantitative polymerase chain reaction; Western blotting; immunohistochjemical analyses; cell proliferation and colony formation assays; cell migration and cell invasion assays) were employed to assess various aspects of UBE4B.We found that UBE4B expression was upregulated aberrantly at mRNA and protein levels in human primary HCC tissues. Amplified expression of UBE4B was highly correlated with poor outcome. Silencing of UBE4B expression by siRNA inhibited the proliferation, colony formation, migration and invasion of HCC cells in vitro, and resulted in significant apoptosis that was associated with downregulation of expression of Bcl-2 and upregulation of expression of total p53, p-p53, Bax and Cleaved-Caspase3 in HCC cells. Our findings suggested that UBE4B might have an oncogenic role in human primary HCC, and that it could be used as a prognostic marker (as well as a potential molecular target) for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Gene Expression , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Adult , Aged , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Cell Proliferation , Female , Gene Knockdown Techniques , Gene Silencing , Humans , Immunohistochemistry , Liver Neoplasms/diagnosis , Liver Neoplasms/surgery , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Risk Factors , Tumor Burden , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases
3.
Stem Cells ; 33(2): 354-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25267273

ABSTRACT

Cancer stem-like cells/cancer-initiating cells (CSCs/CICs) are considered to represent a small population of cancer cells that is resistant to conventional cancer treatments and responsible for tumor recurrence and metastasis. The aim of this study was to establish CSC/CIC-targeting immunotherapy. In this study, we found that Annexin A3 (ANXA3) was preferentially expressed in CSCs/CICs derived from hepatocellular carcinoma (HCC) cells compared to non-CSCs/CICs. In HCC samples, high levels of ANXA3 correlated with expansion of CD133(+) tumor cells representing CSCs/CICs in HCC; the combination of high levels of ANXA3 and CD133 was associated with progression of HCC. Overexpression of ANXA3 increased the proportion of CD133(+) cells, enhancing their tumorigenicity. On the contrary, knockdown of ANXA3 decreased CD133(+) cells and inhibited tumorigenicity. The mechanistic study revealed that ANXA3-mediated maintenance of HCC CSCs/CICs activity was likely involved with the HIF1A/Notch pathway. Using ANXA3 as a target, ANXA3-transfected dendritic cells could induce more functionally active T cells and these effector T cells could superiorly kill CD133(+) HCC CSCs/CICs in vitro and in vivo. Taken together, our findings suggest that ANXA3 plays a role in HCC CSC/CIC maintenance, and that ANXA3 may represent a potential CSC/CIC-specific therapeutic target for improving the treatment of HCC.


Subject(s)
Annexin A3/immunology , Immunotherapy , Liver Neoplasms/therapy , Neoplasm Proteins/immunology , Neoplastic Stem Cells/immunology , AC133 Antigen , Animals , Annexin A3/genetics , Antigens, CD/genetics , Antigens, CD/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Peptides/genetics , Peptides/immunology , Receptors, Notch/genetics , Receptors, Notch/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transfection
4.
J Transl Med ; 12: 273, 2014 Sep 27.
Article in English | MEDLINE | ID: mdl-25260879

ABSTRACT

BACKGROUND: Galectin-3, a member of the beta-galactoside-binding lectin family, is a multifunctional protein with various biological functions, including the proliferation and differentiation of tumor cells, angiogenesis, cancer progression, and metastasis. We aimed to clarify if expression of galectin-3 is related to the clinicopathological characteristics and prognosis of hepatocellular carcinoma (HCC) patients, and to explore the possible mechanisms of galectin-3 in hepatocellular carcinoma. METHODS: First, we investigated galectin-3 mRNA and protein expression by using RT-PCR and Western blotting. Second, tissues from 165 HCC patients were used to evaluate clinicopathological characteristics and prognosis through immunohistochemical analyses. Furthermore, the functions of galectin-3 were analyzed with respect to the proliferation, cell cycle,apoptosis, migration, and invasion of HCC cell lines. Finally, we analyzed galectin-3 expression and micro-vessel density (MVD) by immunohistochemistry (IHC) to find its correlation with angiogenesis in Hepatocellular Carcinoma. Flow cytometer was used to explore apoptosis and Western-blot was used to detect the pathway proteins of apoptosis. RESULTS: Galectin-3 showed high expression at the mRNA and protein levels in HCC cancer tissues and cell lines. Clinicopathological analyses revealed that increased expression of galectin-3 in tumors was closely associated with a poor prognosis. Galectin-3 knockdown by siRNA significantly inhibited cell growth, migration, and invasion, and induced apoptosis in HCC cells in vitro, whereas galectin-3 overexpression promoted cell growth, migration, and invasion. Correlation analysis of galectin-3 expression and micro-vessel density (MVD) showed that galectin-3 expression in tumor cells stimulates angiogenesis. The observed regulation of cell apoptosis was accompanied by the galectin-3-mediated modulation of caspase3 signaling pathways in HCC cells. CONCLUSIONS: These data suggest that galectin-3 plays an important part in HCC progression and may serve as a prognostic factor for HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Galectin 3/metabolism , Liver Neoplasms/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Proliferation , Female , Galectin 3/genetics , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Prognosis
5.
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
6.
Cancer Res ; 84(10): 1613-1629, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38381538

ABSTRACT

Neutrophil extracellular traps (NET), formed by the extracellular release of decondensed chromatin and granules, have been shown to promote tumor progression and metastasis. Tumor-associated neutrophils in hepatocellular carcinoma (HCC) are prone to NET formation, highlighting the need for a more comprehensive understanding of the mechanisms of action of NETs in liver cancer. Here, we showed that DNA of NETs (NET-DNA) binds transmembrane and coiled-coil domains 6 (TMCO6) on CD8+ T cells to impair antitumor immunity and thereby promote HCC progression. TGFß1 induced NET formation, which recruited CD8+ T cells. Binding to NET-DNA inhibited CD8+ T cells function while increasing apoptosis and TGFß1 secretion, forming a positive feedback loop to further stimulate NET formation and immunosuppression. Mechanistically, the N-terminus of TMCO6 interacted with NET-DNA and suppressed T-cell receptor signaling and NFκB p65 nuclear translocation. Blocking NET formation by inhibiting PAD4 induced potent antitumor effects in wild-type mice but not TMCO6-/- mice. In clinical samples, CD8+ T cells expressing TMCO6 had an exhausted phenotype. TGFß1 signaling inhibition or TMCO6 deficiency combined with anti-PD-1 abolished NET-driven HCC progression in vivo. Collectively, this study unveils the role of NET-DNA in impairing CD8+ T-cell immunity by binding TMCO6 and identifies targeting this axis as an immunotherapeutic strategy for blocking HCC progression. SIGNIFICANCE: TMCO6 is a receptor for DNA of NETs that mediates CD8+ T-cell dysfunction in HCC, indicating that the NET-TMCO6 axis is a promising target for overcoming immunosuppression in liver cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Extracellular Traps , Liver Neoplasms , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Humans , Mice , Extracellular Traps/immunology , Extracellular Traps/metabolism , Transforming Growth Factor beta1/metabolism , Neutrophils/immunology , Neutrophils/metabolism , DNA/immunology , DNA/metabolism , Mice, Inbred C57BL , Mice, Knockout , Cell Line, Tumor , Male
7.
Signal Transduct Target Ther ; 9(1): 79, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38565886

ABSTRACT

Fluoropyrimidine-based combination chemotherapy plus targeted therapy is the standard initial treatment for unresectable metastatic colorectal cancer (mCRC), but the prognosis remains poor. This phase 3 trial (ClinicalTrials.gov: NCT03950154) assessed the efficacy and adverse events (AEs) of the combination of PD-1 blockade-activated DC-CIK (PD1-T) cells with XELOX plus bevacizumab as a first-line therapy in patients with mCRC. A total of 202 participants were enrolled and randomly assigned in a 1:1 ratio to receive either first-line XELOX plus bevacizumab (the control group, n = 102) or the same regimen plus autologous PD1-T cell immunotherapy (the immunotherapy group, n = 100) every 21 days for up to 6 cycles, followed by maintenance treatment with capecitabine and bevacizumab. The main endpoint of the trial was progression-free survival (PFS). The median follow-up was 19.5 months. Median PFS was 14.8 months (95% CI, 11.6-18.0) for the immunotherapy group compared with 9.9 months (8.0-11.8) for the control group (hazard ratio [HR], 0.60 [95% CI, 0.40-0.88]; p = 0.009). Median overall survival (OS) was not reached for the immunotherapy group and 25.6 months (95% CI, 18.3-32.8) for the control group (HR, 0.57 [95% CI, 0.33-0.98]; p = 0.043). Grade 3 or higher AEs occurred in 20.0% of patients in the immunotherapy group and 23.5% in the control groups, with no toxicity-associated deaths reported. The addition of PD1-T cells to first-line XELOX plus bevacizumab demonstrates significant clinical improvement of PFS and OS with well tolerability in patients with previously untreated mCRC.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Oxaloacetates , Humans , Bevacizumab/therapeutic use , Capecitabine/therapeutic use , Oxaliplatin , Colorectal Neoplasms/drug therapy , Fluorouracil/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Immunotherapy
8.
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
9.
Chin J Cancer ; 32(10): 553-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23470144

ABSTRACT

Chuankezhi (CKZ), a new Chinese medicine, plays an important role in immunoregulation. Cytokine-induced killer (CIK) cells have been commonly used for immunotherapy in recent years. In this study, we aimed to investigate the immunoregulatory effect of CKZ on CIK cells. Peripheral blood monocytes were isolated from healthy donors, and CIK cells were generated by culturing monocytes with interferon-gamma (IFN-γ) and interleukin 2. Different concentrations of CKZ were added on day 2. After incubation for 14 days in culture, the antitumor effects of CIK cells were measured by cytotoxicity assay. Flow cytometry was used to explore the effect of CKZ on CIK cell immunophenotype, intracellular cytokine production, and apoptosis. The effect of CKZ on the antitumor activity of CIK cells in nude mice was also investigated. CKZ increased the percentage of CD3+CD56+ CIK cells but did not significantly change the percentage of CD4+, CD8+, or CD4+CD25+ CIK cells. CKZ-conditioned CIK cells showed a greater ability to kill tumor cells, as well as a higher frequency of IFN-γ and TNF-α production, compared with the CIK cells in the control group. CKZ also suppressed the apoptosis of CIK cells in vitro. Furthermore, CKZ combined with CIK cells had a stronger suppressive effect on tumor growth in vivo than the CIK, CKZ, or normal saline control groups. Our results indicate that CKZ enhances the antitumor activity of CIK cells and is a potential medicine for tumor immunotherapy.


Subject(s)
Apoptosis , Cytokine-Induced Killer Cells/immunology , Drugs, Chinese Herbal/pharmacology , Epimedium/chemistry , Morinda/chemistry , Animals , Apoptosis/drug effects , CD3 Complex/metabolism , CD56 Antigen/metabolism , Cell Line, Tumor/drug effects , Cytokine-Induced Killer Cells/cytology , Cytokine-Induced Killer Cells/drug effects , Cytotoxicity, Immunologic , Drugs, Chinese Herbal/isolation & purification , Female , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Plants, Medicinal/chemistry , Tumor Burden , Tumor Necrosis Factor-alpha/metabolism
10.
Chin J Cancer ; 32(12): 661-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23706769

ABSTRACT

In vitro amplified human leukocyte antigen (HLA)-haploidentical donor immune cell infusion (HDICI) is not commonly used in children. Therefore, our study sought to evaluate its safety for treating childhood malignancies. Between September 2011 and September 2012, 12 patients with childhood malignancies underwent HDICI in Sun Yat-sen University Cancer Center. The median patient age was 5.1 years (range, 1.7-8.4 years). Of the 12 patients, 9 had high-risk neuroblastoma (NB) [7 showed complete response (CR), 1 showed partial response (PR), and 1 had progressive disease (PD) after multi-modal therapies], and 3 had Epstein-Barr virus (EBV)-positive lymphoproliferative disease (EBV-LPD). The 12 patients underwent a total of 92 HDICIs at a mean dose of 1.6×10(8) immune cells/kg body weight: 71 infusions with natural killer (NK) cells, 8 with cytokine-induced killer (CIK) cells, and 13 with cascade primed immune cells (CAPRIs); 83 infusions with immune cells from the mothers, whereas 9 with cells from the fathers. Twenty cases (21.7%) of fever, including 6 cases (6.5%) accompanied with chills and 1 (1.1%) with febrile convulsion, occurred during infusions and were alleviated after symptomatic treatments. Five cases (5.4%) of mild emotion changes were reported. No other adverse events occurred during and after the completion of HDIDIs. Neither acute nor chronic graft versus host disease (GVHD) was observed following HDICIs. After a median of 5.0 months (range, 1.0-11.5 months) of follow-up, the 2 NB patients with PR and PD developed PD during HDICIs. Of the other 7 NB patients in CR, 2 relapsed in the sixth month of HDICIs, and 5 maintained CR with disease-free survival (DFS) ranging from 4.5 to 11.5 months (median, 7.2 months). One EBV-LPD patient achieved PR, whereas 2 had stable disease (SD). Our results show that HDICI is a safe immunotherapy for childhood malignancies, thus warranting further studies.


Subject(s)
Cytokine-Induced Killer Cells/immunology , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Lymphoproliferative Disorders/therapy , Neuroblastoma/therapy , Child , Child, Preschool , Epstein-Barr Virus Infections/therapy , Female , Follow-Up Studies , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunotherapy, Adoptive , Infant , Lymphoproliferative Disorders/virology , Male , Transplantation, Homologous , Treatment Outcome
11.
J Gastroenterol Hepatol ; 26(3): 544-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21332550

ABSTRACT

BACKGROUND AND AIM: Exportin 4 (XPO4) is a recently-discovered candidate tumor-suppressor gene identified in a liver cancer mouse model. To investigate the role of XPO4 in hepatocellular carcinoma (HCC) pathogenesis, we determined XPO4 expression and its correlation to prognosis in human primary HCC. METHODS: The XPO4 mRNA transcription level in HCC cell lines and tissue samples were detected by real-time quantitative polymerase chain reaction (PCR). XPO4 protein expression in 123 primary HCC clinical surgical specimens were analyzed by immunohistochemical detection. RESULTS: Real-time quantitative PCR showed a decrease in XPO4 expression in HCC cell lines BEL-7402, Hep-G2, and SK-hep1 compared to the normal liver cell line LO2. Decreased XPO4 mRNA was also found in the majority of tumor tissues compared with matched non-tumor liver tissues (P = 0.004). Immunohistochemical detection revealed that XPO4 expression was reduced in 51 of 123 (41.5%) tumor resection samples compared with adjunct non-tumor tissues. We also found XPO4 expression to be significantly correlated with tumor size (P = 0.045) and histopathological classification (P = 0.004). Kaplan-Meier survival curves showed that the downregulation of XPO4 resulted in a significantly poor prognosis (P = 0.008, log-rank test), and multivariate Cox's analysis showed that XPO4 expression was an independent prognostic factor for overall survival of HCC patients (P = 0.013). CONCLUSIONS: Our data suggest that XPO4 could be involved in the progression of human HCC and could serve as a potential target for gene therapy in the treatment of HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Karyopherins/metabolism , Liver Neoplasms/metabolism , Adult , Aged , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Chi-Square Distribution , China , Down-Regulation , Female , Hep G2 Cells , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Karyopherins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Risk Assessment , Risk Factors , Survival Rate , Time Factors , Young Adult
12.
Clin Transl Immunology ; 10(3): e1257, 2021.
Article in English | MEDLINE | ID: mdl-33717483

ABSTRACT

OBJECTIVES: Although axitinib has achieved a preferable response rate for advanced renal cell carcinoma (RCC), patient survival remains unsatisfactory. In this study, we evaluated the efficacy and safety of a combination treatment of axitinib and a low dose of pembrolizumab-activated autologous dendritic cells-co-cultured cytokine-induced killer cells in patients with advanced RCC. METHODS: All adult patients, including treatment-naive or pretreated with VEGF-targeted agents, were enrolled from May 2016 to March 2019. Patients received axitinib 5 mg twice daily and pembrolizumab-activated dendritic cells-co-cultured cytokine-induced killer cells intravenously weekly for the first four cycles, every 2 weeks for the next four cycles, and every month thereafter. RESULTS: The 43 patients (22 untreated and 21 previously treated) showed a median progression-free survival (mPFS) of 14.7 months (95% CI, 11.16-18.30). mPFS in treatment-naive patients was 18.2 months, as compared with 14.4 months in pretreated patients (log-rank P-value = 0.07). Overall response rates were 25.6% (95% CI, 13.5-41.2%). Grade 3 or higher adverse events occurred in 5% of patients included hypertension (11.6%) and palmar-plantar erythrodysesthesia (7.0%). Peripheral blood lymphocyte immunophenotype and serum cytokine profile analyses demonstrated increased antitumor immunity after combination treatment particularly in patients with a long-term survival benefit, while those with a minimal survival benefit demonstrated an elevated proportion of peripheral CD8+TIM3+ T cells and lower serum-level immunostimulatory cytokine profile. CONCLUSIONS: The combination therapy was active and well tolerated for treatment of advanced RCC, either as first- or second-line treatment following other targeted agents. Changes in immunophenotype and serum cytokine profile may be used as prognostic biomarkers.

13.
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
14.
Clin Transl Immunology ; 9(2): e1113, 2020.
Article in English | MEDLINE | ID: mdl-32076550

ABSTRACT

OBJECTIVES: Fluoropyrimidine-based chemotherapy regimens are the current first-line treatment for metastatic colorectal cancer (mCRC); however, the outcome is often unsatisfactory. The present study aimed to determine the effect of combined cytokine-induced killer (CIK) cell immunotherapy and first-line chemotherapy in patients with mCRC. METHODS: This retrospective study included 252 patients with mCRC treated with first-line chemotherapy. Among them, 126 patients received first-line chemotherapy only (control group), while the other 126 patients, with similar demographic and clinical characteristics, received CIK cell immunotherapy combined with first-line chemotherapy (CIK group). Overall survival (OS) and progression-free survival (PFS) were compared between the two groups using the Kaplan-Meier method. RESULTS: The median OS for the CIK group was 54.7 versus 24.1 months for the controls, and the median PFS for the CIK group was 25.7 versus 14.6 months for the controls. Univariate and multivariate analyses indicated that CIK cell treatment was an independent prognostic factor for patients' OS and PFS. Subgroup analyses showed that CIK cell treatment significantly improved the OS and PFS of patients with metastatic colon cancer, but not those with metastatic rectal cancer. Additionally, the change in CD3+CD56+ subsets after the fourth treatment cycle might be an indicator of successful CIK cell treatment: Patients with increased CD3+CD56+ subsets had better survival than those with decreased CD3+CD56+ subsets. CONCLUSION: Cytokine-induced killer cell immunotherapy combined with first-line chemotherapy could significantly improve the OS and PFS of patients with mCRC, particularly for patients with metastatic colon cancer.

15.
J Mol Endocrinol ; 64(1): 43-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31786540

ABSTRACT

Receptive endometrium is a prerequisite for successful embryo implantation, and it follows that poor endometrial receptivity is a leading cause of implantation failure. miRNAs play important roles as epigenetic regulators of endometrial receptivity and embryo implantation through post-transcriptional modifications. However, the mechanisms of action of many miRNAs are poorly understood. In this study, we investigated the role of the miR-183 family, comprising three miRNAs (miR-183-5p, miR-182-5p, and miR-96-5p) in endometrial receptivity and embryo implantation. The miR-183 family shows estrogen-dependent upregulation in endometrial Ishikawa (IK) cells. The miR-183 family also has a positive role in migration and proliferation of IK cells. Furthermore, JAr spheroid attachment experiments show that attachment rates were significantly decreased after treatment of IK cells with inhibitors for miR-183-5p and miR-182-5p and increased after treatment with miR-183-5p-mimic and miR-96-5p-mimic, respectively. The downstream analysis shows that catenin alpha 2 (CTNNA2) is a potential target gene for miR-183-5p, and this was confirmed in luciferase reporter assays. An in vivo mouse pregnancy model shows that inhibition of miR-183-5p significantly decreases embryo implantation rates and increases CTNNA2 expression. Downregulation of CTNNA2 in endometrial cells by miR-183-5p may be significant in mediating estrogenic effects on endometrial receptivity. In conclusion, miR-183-5p and the CTNNA2 gene may be potential biomarkers for endometrial receptivity and may be useful diagnostic and therapeutic targets for successful embryo implantation.


Subject(s)
Embryo Implantation/genetics , MicroRNAs/genetics , Uterus/physiology , Animals , Biomarkers/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation/genetics , Embryo Implantation/physiology , Endometrium/physiology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred ICR , Pregnancy , alpha Catenin/genetics
16.
Oncoimmunology ; 9(1): 1752563, 2020.
Article in English | MEDLINE | ID: mdl-32363125

ABSTRACT

Adjuvant chemotherapy after surgery is the standard treatment modality for stage III and part of stage II or stage IV colorectal cancer (CRC) patients. However, the 5-year overall survival (OS) rate remains unsatisfactory. Thus, developing combination therapies is essential to improve the prognosis of patients with CRC. The present study aimed to determine the effect of a sequential combination of cytokine-induced killer cell (CIK) infusion and chemotherapy for patients with CRC. 122 patients with CRC treated with postoperative adjuvant chemotherapy were retrospectively included in this study. Among them, 62 patients received adjuvant chemotherapy only (control group), while the other 60 patients, with similar demographic and clinical characteristics, received adjuvant chemotherapy and sequential CIK cell immunotherapy (CIK group). Survival analysis showed significantly improved disease free survival (DFS) and OS rates in the CIK group compared with the control group (log-rank test, P = .0024; P = .008, respectively). Univariate and multivariate analyses indicated that sequential CIK cell treatment was an independent prognostic factor for patients' DFS and OS. Subgroup analyses showed that sequential CIK cell treatment significantly improved the DFS and OS of patients with high-risk T4 stage and insufficient chemotherapy duration. In conclusion, these data indicate that sequential adjuvant CIK cell treatment combined with chemotherapy is an effective therapeutic strategy to prevent disease recurrence and prolong survival of patients with CRC, particularly for patients with high-risk T4 stage and insufficient chemotherapy duration.


Subject(s)
Colorectal Neoplasms , Cytokine-Induced Killer Cells , Chemotherapy, Adjuvant , Colorectal Neoplasms/therapy , Combined Modality Therapy , Digestive System Surgical Procedures , Female , Humans , Immunotherapy, Adoptive , Male , Middle Aged , Neoplasm Recurrence, Local , Retrospective Studies
17.
J Cancer ; 10(5): 1154-1161, 2019.
Article in English | MEDLINE | ID: mdl-30854124

ABSTRACT

Purpose: To evaluate the tumour cell PD-L1 (TC-PD-L1) expression patterns in the local microenvironment of clear cell renal cell carcinoma (ccRCC). Materials and Methods: 30 fresh primary ccRCC tissues were used to detect the association between TC-PD-L1 and CD8+TILs at mRNA level. The in vitro incubation experiment was used to confirm the association between extrinsic TC-PD-L1 expression and IFNγ. A cohort of 135 ccRCC patients treated between January 2009 and August 2013 was included for survival analysis. Results: Our results confirmed that ccRCC cell lines were capable of expressing PD-L1. The incubation experiment in vitro demonstrated the positive correlation of TC-PD-L1 expression with interferon-gamma (IFNγ). Additionally, survival analysis was investigated in 135 ccRCC patients and found no independent correlation of TC-PD-L1 expression in multivariate analysis, whereas more distinct prognostic differences were detected when TC-PD-L1-positive ccRCC were sub-classified as with or without CD8+ T cell infiltration. Conclusion: The intrinsic and extrinsic expression patterns are both detected in ccRCC. High positive rate of TC-PD-L1 correlated closely to the strong infiltration of CD8+ TILs. TC-PD-L1-positive ccRCC patients with abundant CD8+ TILs infiltration confer the high risk of death and disease relapse.

18.
Reprod Toxicol ; 84: 9-17, 2019 03.
Article in English | MEDLINE | ID: mdl-30562550

ABSTRACT

Although most children conceived by assisted reproductive technology (ART) are healthy, there are concerns regarding the potential long-term health implications of ART. It has been reported that alterations in insulin-induced gene (INSIG), sterol regulatory element binding protein (SREBP), and SREBP cleavage-activating protein (SCAP) are involved in cardiometabolic changes. Thus, ART mouse models were established via in vitro fertilization (IVF), intracytoplasmic injection (ICSI), and in vitro oocyte maturation (IVM). A significantly higher systolic blood pressure was identified in the IVM aged female mice. In addition, abnormalities in the blood lipids and liver function were identified in the IVM- or ICSI-conceived elderly mice. Furthermore, ICSI or IVM significantly affected the hepatic expression and methylation of INSIG-SCAP-SREBP from a young to old age. Our animal data indicated that ICSI or IVM result in a higher risk of cholesterol metabolism dysfunction in older mice, which may be associated with long-term alterations of INSIG-SCAP-SREBP.


Subject(s)
Aging/physiology , Intracellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Membrane Proteins/genetics , Reproductive Techniques, Assisted , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Animals , Blood Pressure , Cholesterol/metabolism , Female , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Pregnancy
19.
Oncoimmunology ; 7(4): e1417721, 2018.
Article in English | MEDLINE | ID: mdl-29632736

ABSTRACT

Cytokine-induced killer (CIK) cells that are stimulated using mature dendritic cells (DCs), referred to as (DC-CIK cells) exhibit superior anti-tumor potency. Anti-programmed death-1 (PD-1) antibodies reinvigorate T cell-mediated antitumor immunity. This phase I study aimed to assess the safety and clinical activity of immunotherapy with PD-1 blockade (pembrolizumab)-activated autologous DC-CIK cells in patients with advanced solid tumors. Patients with selected types of advanced solid tumors received a single intravenous infusion of activated autologous DC-CIK cells weekly for the first month and every 2 weeks thereafter. The primary end points were safety and adverse event (AE) profiles. Antitumor responses, overall survival (OS), progression-free survival (PFS) and cytolytic activity were secondary end points. Treatment-related AEs occurred in 20/31 patients. Grade 3 or 4 toxicities, including fever and chills, were observed in two patients. All treatment-related AEs were reversible or controllable. The cytotoxicity of DC-CIK cells induced up-regulation of PD-L1 expression on autologous tumor cells. When activated using pembrolizumab ex vivo, DC-CIK cells exerted superior antitumor properties and elevated IFN-γ secretion. Objective responses (complete or partial responses) were observed in 7 of the 31patients.These responses were durable, with 6 of 7 responses lasting more than 5 months. The overall disease control rate in the patients was 64.5%. At the time of this report, the median OS and PFS were 270 and 162 days, respectively. In conclusions, treatment with pembrolizumab-activated autologous DC-CIK cells was safe and exerted encouraging antitumor activity in advanced solid tumors. A larger phase II trial is warranted.

20.
Zhonghua Zhong Liu Za Zhi ; 29(6): 411-4, 2007 Jun.
Article in Zh | 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
SELECTION OF CITATIONS
SEARCH DETAIL