Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 113(5): 747-55, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26284335

ABSTRACT

BACKGROUND: Tumoural infiltration of T lymphocytes is determined by local patterns of specific chemokine expression. In this report, we examined the roles of CCL4 and CCL20 in the accumulation of CD8(+) cytotoxic T lymphocytes (CTLs) and regulatory T (Treg) lymphocytes in oesophageal squamous cell carcinoma (ESCC), and determined the correlations between chemokine expressions and ESCC patients' survival. METHODS: Reverse transcriptase-PCR and immunohistochemistry (IHC) staining were performed to examine the expressions of interested genes. Flow cytometry was adopted to check the expressions of CCL4- and CCL6-specific receptors, CCR5 and CCR6, on CTLs and Treg cells. In addition, transwell assay was carried on. RESULTS: The CCL4 expression was significantly correlated with the expression of CTL markers (CD8 and Granzyme B), whereas CCL20 was positively correlated with Treg markers (FoxP3 and IL-10). Consistently, CCR5 was found to be mainly expressed on CD8(+) T lymphocytes, while CCR6 showed prevalence on Treg lymphocytes and the frequencies of CCR5(+)CD8(+) CTLs and CCR6(+) Treg cells were higher in TIL compared with PBMC. Respectively, CCL4 and CCL20 recruited CD8(+) and regulatory T cells in vitro. Importantly, high levels of CCL4 in the lesions of ESCC patients predicted prolonged survival. Furthermore, CCL4(high)/CCL20(low) group demonstrated better overall survival, whereas CCL4(low)/CCL20(low) and CCL4(low)/CCL20(high) groups showed the worst overall survival. CONCLUSIONS: Our data showed that CCL4 and CCL20 recruit functionally different T lymphocyte subsets into oesophageal carcinoma, indicating CCL4 and CCL20 are potential predictors of ESCC patients' survival.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Chemokine CCL20/metabolism , Chemokine CCL4/metabolism , Esophageal Neoplasms/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/secondary , Cell Movement , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Multivariate Analysis , Prognosis , Proportional Hazards Models , Receptors, CCR5/metabolism , Receptors, CCR6/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology
2.
Br J Cancer ; 110(8): 1992-9, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24642619

ABSTRACT

BACKGROUND: Cytokine-induced killer (CIK) cells are ex vivo-expanded immune cells that express NK-cell and T-cell markers and that are routinely used in the treatment of many cancers. One key advantage of CIK cells is their ability to efficiently traffic to many solid tumours. Although likely to be mediated by chemokine receptor (CKR) expression, a thorough examination of the mechanism of tumour targeting has not been previously explored. METHODS: Here, human CIK cell expansions were examined for the level, profile and kinetics of CKR expression. RESULTS: It was found that CIK cells express a panel of CKRs, with considerable variation between donors. Importantly, CKR levels dropped considerably beyond 14 days in culture, being significantly reduced by day 28 (the time at which cytolytic activity peaked). As such, CIK preparations that are used clinically may not have optimal CKR expression. Several approaches were found to re-stimulate CKR cell-surface levels at these later time points. These approaches also enhanced cytolytic activity in vitro and were demonstrated to increase both in vivo tumour trafficking and anti-tumour activity in mouse models. CONCLUSIONS: Simple modifications of the CIK expansion protocol could therefore be used to significantly enhance the anti-tumour effects of this therapy.


Subject(s)
Cytokine-Induced Killer Cells/metabolism , Cytotoxicity, Immunologic , Killer Cells, Natural/metabolism , Neoplasms/immunology , Receptors, Chemokine/metabolism , Animals , Cell Proliferation , Cytokine-Induced Killer Cells/immunology , Flow Cytometry , HeLa Cells , Humans , Killer Cells, Natural/immunology , Mice , Neoplasms/therapy , Receptors, Chemokine/immunology
3.
Gene Ther ; 20(7): 770-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23282955

ABSTRACT

Despite significant strides made in the clinical translation of adoptive immune cell therapies, it is apparent that many tumors incorporate strategies to avoid recognition by receptors expressed on the immune cells, such as NKG2D. Strategies that stabilize the expression of ligands for these receptors may enhance the therapeutic potential of these and related therapies. Doxycycline inhibits matrix metalloproteinases (MMPs) that act to cleave the extracellular domain of MICA/B, ligands for the NKG2D receptor. Doxycycline treatment blocked shedding of MICA/B from a panel of human tumor cells, but also acted to increase their expression and cell surface translocation, possibly through its action on ATM. This meant that many tumor cells displayed increased MICA/B expression and enhanced susceptibility to CIK cells. Interestingly, doxycycline also selectively enhanced the replication of oncolytic vaccinia in many tumor cell lines, leading to increased sensitivity to these therapies. Combination (CIK-oncolytic vaccinia) therapies used in conjunction with doxycycline led to increased anti-tumor effects. The unexpected and pleiotropic beneficial anti-tumor effects of doxycycline on both immune cell and oncolytic viral therapies make it an excellent candidate for rapid clinical testing.


Subject(s)
Doxycycline/administration & dosage , Immunotherapy, Adoptive , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms/drug therapy , Oncolytic Virotherapy , Cell Line, Tumor , Combined Modality Therapy , Cytotoxicity, Immunologic , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasms/genetics , Neoplasms/pathology , Translational Research, Biomedical , Vaccinia virus/genetics
4.
Oncogene ; 30(3): 323-33, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20856202

ABSTRACT

Metastasis is the major reason for breast cancer-related deaths. Although there is a host of indirect evidence for a role of protein kinase C (PKC) α in primary breast cancer growth, its role in the molecular pathways leading to metastasis has not been studied comprehensively. By treating mice with αV5-3, a novel peptide inhibitor selective for PKCα, we were able to determine how PKCα regulates metastasis of mammary cancer cells using a syngeneic and orthotopic model. The primary tumor growth was not affected by αV5-3 treatment. However, the mortality rate was reduced and metastasis in the lung decreased by more than 90% in the αV5-3-treated mice relative to the control-treated mice. αV5-3 treatment reduced intravasation by reducing matrix metalloproteinase-9 activities. αV5-3 treatment also reduced lung seeding of tumor cells and decreased cell migration, effects that were accompanied by a reduction in nuclear factor kappa B activity and cell surface levels of the CXCL12 receptor, CXCR4. αV5-3 treatment caused no apparent toxicity in non-tumor-bearing naïve mice. Rather, inhibiting PKCα protected against liver damage and increased the number of immune cells in tumor-bearing mice. Importantly, αV5-3 showed superior efficacy relative to anti-CXCR4 antibody in reducing metastasis in vivo. Together, these data show that pharmacological inhibition of PKCα effectively reduces mammary cancer metastasis by targeting intravasation and lung seeding steps in the metastatic process and suggest that PKCα-specific inhibitors, such as αV5-3, can be used to study the mechanistic roles of PKCα specifically and may provide a safe and effective treatment for the prevention of lung metastasis of breast cancer patients.


Subject(s)
Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis , Neoplasm Seeding , Protein Kinase C-alpha/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , RNA, Small Interfering , Receptors, CXCR4/metabolism
5.
Gene Ther ; 18(2): 164-72, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20739958

ABSTRACT

Recent developments in the field of oncolytic or tumor-selective viruses have meant that the clinical applications of these agents are now being considered in more detail. Like most cancer therapies it is likely that they will be used primarily in combination with other therapeutics. Although several reports have shown that oncolytic viruses can synergize with chemotherapies within an infected cancer cell, it would be particularly important to determine whether factors released from infected cells could enhance the action of chemotherapies at a distance. Here, we demonstrate in vitro synergy between oncolytic vaccinia and taxanes. However, we also show, for the first time, that this synergy is at least partly due to the release of factors from the infected cells that are capable of sensitizing surrounding cells to chemotherapy. Several cellular factors were identified as being mediators of this bystander effect, including type I interferon released soon after infection and high-mobility group protein B1 (HMGB1) released after cell death. This represents the first description of these mechanisms for beneficial interactions between viral and traditional tumor therapies. These data may provide a direct basis for the design of clinical trials with agents currently in the clinic, as well as providing insight into the development of next generation viral vectors.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , HMGB1 Protein/metabolism , Interferons/metabolism , Neoplasms/therapy , Oncolytic Virotherapy/methods , Paclitaxel/therapeutic use , Vaccinia virus , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Mice , Mice, Nude , Oncolytic Viruses/physiology , Paclitaxel/pharmacology , Vaccinia virus/physiology
6.
Gene Ther ; 15(10): 753-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18356814

ABSTRACT

Despite significant advances in the development of tumor-selective agents, strategies for effective delivery of these agents across biological barriers to cells within the tumor microenvironment has been limiting. One tactical approach to overcoming biological barriers is to use cells as delivery vehicles, and a variety of different cell types have been investigated with a range of agents. In addition to transporting agents with targeted delivery, cells can also produce their own tumoricidal effect, conceal a payload from an immune response, amplify a selective agent at the target site and facilitate an antitumor immune response. We have reported a therapeutic combination consisting of cytokine induced killer cells and an oncolytic vaccinia virus with many of these features that led to therapeutic synergy in animal models of human cancer. The synergy was due to the interaction of the two agents to enhance the antitumor benefits of each individual component. As both of these agents display broad tumor-targeting potential and possess unique tumor killing mechanisms, together they were able to recognize and destroy a far greater number of malignant cells within the heterogeneous tumor than either agent alone. Effective cancer therapy will require recognition and elimination of the root of the disease, the cancer stem cell, and the combination of CIK cells and oncolytic vaccinia viruses has this potential. To create effective tumor-selective agents the viruses are modified to take advantage of the unique biology of the cancer cell. Similarly, if we are to develop targeted therapies that are sufficiently multifaceted to eliminate cancer cells at all stages of disease, we should integrate the virus into the unique biology of the cell delivery vehicle.


Subject(s)
Genetic Therapy/methods , Killer Cells, Natural/transplantation , Neoplasms/therapy , Oncolytic Virotherapy/methods , Animals , Combined Modality Therapy , Cytokines/immunology , Genetic Engineering , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Neoplasms/immunology , Vaccinia virus/physiology
7.
Cell Mol Life Sci ; 64(12): 1449-51, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17404689

ABSTRACT

A variety of viral-based and immune cell therapies have been proposed for use in the treatment of cancer. One possible approach to improve the effectiveness of these biological agents may be to combine them such that we can take advantage of natural immune cell-pathogen relationships. Here we discuss these potential approaches with particular emphasis on the use of immune cells as carrier vehicles to deliver viral therapies to the tumor.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , Oncolytic Virotherapy , Animals , Combined Modality Therapy
8.
Mol Ther ; 14(3): 361-70, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16905462

ABSTRACT

Targeted oncolytic viruses and immunostimulatory therapeutics are being developed as novel cancer treatment platforms. These approaches can be combined through the expression of immunostimulatory cytokines from targeted viruses, including adenoviruses and herpesviruses. Although intratumoral injection of such viruses has been associated with tumor growth inhibition, eradication of distant metastases was not reported. The major limitations for this approach to date have been (1) inefficient intravenous virus delivery to tumors and (2) the lack of predictive, immunocompetent preclinical models. To overcome these hurdles, we developed JX-594, a targeted, thymidine kinase(-) vaccinia virus expressing human GM-CSF (hGM-CSF), for intravenous (i.v.) delivery. We evaluated two immunocompetent liver tumor models: a rabbit model with reproducible, time-dependent metastases to the lungs and a carcinogen-induced rat liver cancer model. Intravenous JX-594 was well tolerated and had highly significant efficacy, including complete responses, against intrahepatic primary tumors in both models. In addition, whereas lung metastases developed in all control rabbits, none of the i.v. JX-594-treated rabbits developed detectable metastases. Tumor-specific virus replication and gene expression, systemically detectable levels of hGM-CSF, and tumor-infiltrating CTLs were also demonstrated. JX-594 holds promise as an i.v.-delivered, targeted virotherapeutic. These two tumor models hold promise for the optimization of this approach.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Liver Neoplasms/therapy , Lung Neoplasms/prevention & control , Oncolytic Virotherapy , Vaccinia virus/genetics , Animals , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Humans , Immunotherapy/methods , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred Strains , Nitrosoguanidines/toxicity , Poxviridae/genetics , Rabbits , Rats , Rats, Sprague-Dawley , T-Lymphocytes, Cytotoxic/immunology , Thymidine Kinase/genetics , Tumor Cells, Cultured , Virus Replication
9.
J Bacteriol ; 181(3): 981-90, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9922264

ABSTRACT

The cell density dependence of stationary-phase survival of Rhizobium leguminosarum has been investigated. Following starvation by exhaustion of carbon or nitrogen, but not of phosphorus, the survival of cultures was dependent on the cell density at entry into stationary phase. High-density cultures survived with little or no loss of viability over a 20-day period in stationary phase. In contrast, low-density cultures lost viability rapidly but consisted of a heterogeneous population, a small fraction of which successfully adapted and eventually formed a stable, surviving population. The threshold density above which the cultures survived successfully in stationary phase was dependent on the growth conditions and the strain used. We took advantage of the fact that R. leguminosarum survives poorly following starvation by resuspension in carbon-free medium to demonstrate that cell density-dependent survival was mediated by a component accumulating in the growth medium. The effects of this medium component on survival in resuspension assays could be mimicked by an N-acyl homoserine lactone, N-(3R-hydroxy-7-cis-tetradecanoyl)-L-homoserine lactone, previously demonstrated to have a role in controlling cell density-dependent phenomena in R. leguminosarum. The Sym plasmids pRP2JI and pRL1JI were found to be essential for the production of the extracellular factor, which could also be made in Escherichia coli carrying the cosmid clone pIJ1086 containing a specific region of pRL1JI.


Subject(s)
Homoserine/analogs & derivatives , Rhizobium leguminosarum/physiology , Colony Count, Microbial , Culture Media , Homeostasis , Homoserine/metabolism , Homoserine/pharmacology , Kinetics , Plasmids , Rhizobium leguminosarum/drug effects , Rhizobium leguminosarum/growth & development , Time Factors
10.
J Bacteriol ; 179(22): 6894-901, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9371432

ABSTRACT

The nitrogen-fixing bacterium Rhizobium leguminosarum bv. phaseoli often has to survive long periods of starvation in the soil, when not in a useful symbiotic relationship with leguminous plants. We report that it can survive carbon, nitrogen, and phosphorus starvation for at least 2 months with little loss of viability. Upon carbon starvation, R. leguminosarum cells were found to undergo reductive cell division. During this period, they acquired the potential for long-term starvation-survival, levels of protein, DNA, and RNA synthesis were decreased to base levels, and pool mRNA was stabilized. The starved cells are ready to rapidly restart growth when nutrients become available. Upon addition of fresh nutrients, there is an immediate increase in the levels of macromolecular synthesis, pool mRNA destabilizes, and the cultures enter exponential growth within 5 to 8 h. The starved cells were cross-protected against pH, heat, osmotic, and oxidative shock. These results provide evidence for a general starvation response in R. leguminosarum similar to that previously found in other bacteria such as Escherichia coli and Vibrio sp.


Subject(s)
Carbon/metabolism , Nitrogen/metabolism , Phosphorus/metabolism , Rhizobium leguminosarum/metabolism , Starvation/metabolism , Bacterial Proteins/metabolism , Culture Media/metabolism , DNA, Bacterial/metabolism , Heating , Hydrogen-Ion Concentration , Kinetics , Macromolecular Substances , Osmosis , Oxidative Stress , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Rhizobium leguminosarum/cytology , Rhizobium leguminosarum/growth & development , S Phase , Survival
SELECTION OF CITATIONS
SEARCH DETAIL
...