Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.029
Filter
1.
BMC Cancer ; 20(1): 678, 2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32689954

ABSTRACT

BACKGROUND: More favorable treatment against epithelial ovarian cancer (EOC) is urgently needed because of its insidious nature at an early stage and a low rate of five-year survival. The current primary treatment, extensive surgery combined with chemotherapy, exhibits limited benefits for improving prognosis. Chimeric antigen receptor T (CAR-T) cell technology as novel immunotherapy has made breakthrough progress in the treatment of hematologic malignancies, and there were also benefits shown in a partial solid tumor in previous research. Therefore, CAR-T cell technology may be a promising candidate as an immunotherapeutic tool against EOC. However, there are some weaknesses in targeting one antigen from the previous preclinical assay, such as on-target off-tumor cytotoxicity. The dual-target CAR-T cell may be a better choice. METHODS: We constructed tandem PD1-antiMUC16 dual-CAR, PD1 single-CAR, and anti-MUC16 single-CAR fragments by PCR and genetic engineering, followed by preparing CAR-T cells via lentiviral infection. The expression of CAR molecules on single and dual CAR-T cells was detected by flow cytometry. The killing capacity and activation of CAR-T cells were measured by cytotoxic assays and cytokines release assays in vitro. The therapeutic capacity of CAR-T cells was assessed by tumor-bearing mice model assay in vivo. RESULTS: We successfully constructed CARs lentiviral expression vectors and obtained single and dual CAR-T cells. CAR-T cells demonstrated robust killing capacity against OVCAR-3 cells in vitro. Meanwhile, CAR-T cells released plenty of cytokines such as interleukin-2(IL-2), interferon-γ (IFN-γ) and tumor necrosis factor-α(TNF-α). CAR-T cells showed a therapeutic benefit against OVCAR-3 tumor-bearing mice and significantly prolonged the survival time. Dual CAR-T cells were shown to be two to four times more efficacious than single CAR-T cells in terms of survival time. CONCLUSION: Although exhibiting a similar ability as single CAR-T cells against OVCAR-3 cells in vitro, dual CAR-T cells demonstrated enhanced killing capacity against OVCAR-3 cells as compared to single CAR-T cells in vivo and significantly prolonged the survival time of tumor-bearing mice. PD1-antiMUC16 CAR-T cells showed more potent antitumor activity than single CAR-T cells in vivo. The present experimental data may support further research work that will have the potential to lead to clinical studies.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Ovarian Epithelial/therapy , Immunotherapy, Adoptive/methods , Membrane Proteins/antagonists & inhibitors , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Animals , B7-H1 Antigen/immunology , CA-125 Antigen/immunology , Carcinoma, Ovarian Epithelial/immunology , Female , Heterografts , Interferon-gamma/metabolism , Interleukin-2/metabolism , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Lentivirus , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Membrane Proteins/immunology , Mice , Neoplasm Transplantation , Ovarian Neoplasms/immunology , Random Allocation , Receptors, Chimeric Antigen/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
J Immunol Methods ; 477: 112690, 2020 02.
Article in English | MEDLINE | ID: mdl-31678265

ABSTRACT

Adoptive cell therapy (ACT) using tumor antigen-independent killer cells has been widely used in clinical trials of cancer treatment. Circumventing the need for identification of a particular tumor-associated antigen on tumor cells, the approach has opened possibilities for the extension of ACT immunotherapy to patients with a wide variety of cancer types. Namely, Natural Killer (NK), Lymphokine-activated Killer (LAK) cells and Cytokine-induced killer (CIK) cells are the most commonly used cell types in antigen-independent adoptive immunotherapy of cancer. They all originate from peripheral blood mononuclear cells and share several common features in their killing mechanisms. However, despite broad application in clinical settings, the boundaries between these cell types are not very clearly defined. The current study aims to review different aspects of these cell populations in terms of phenotypical characteristic and preparation media, to clarify how the boundaries are set.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Primary Cell Culture/methods , Clinical Trials as Topic , Culture Media , Cytokine-Induced Killer Cells/immunology , History, 20th Century , History, 21st Century , Humans , Immunotherapy, Adoptive/history , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Treatment Outcome
3.
Cancer Immunol Res ; 7(11): 1742-1747, 2019 11.
Article in English | MEDLINE | ID: mdl-31676567

ABSTRACT

Natural killer (NK) cells are critical effector lymphocytes mediating tumor immune surveillance and clearance. They do so by direct tumor killing using cytolytic granules and death receptors, and by interfacing with and potentiating adaptive immune responses through the production of cytokines. From a therapeutic perspective, NK cells have been shown to exert graft-versus-leukemia activity in the context of hematopoietic stem cell transplantation and are important in the clinical efficacy of antibodies. Advances in basic and translational NK cell biology have led to multiple potential strategies to augment their in vivo activity to improve antitumor responses. Despite their potent effects, NK cells have been shown to be safe for adoptive cell therapy in both the autologous and allogeneic settings, with promising, but so far limited, clinical efficacy. This review will provide an overview of strategies being pursued to improve NK cell activity and efficacy, focusing on cell source, NK cell activation, and in vivo persistence.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Neoplasms/therapy , Animals , Cell Survival , Cytokines/immunology , Cytokines/therapeutic use , Humans , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/transplantation , Killer Cells, Natural/cytology , Lymphocyte Activation , Neoplasms/immunology , Tumor Microenvironment
4.
Curr Mol Med ; 18(5): 335-342, 2018.
Article in English | MEDLINE | ID: mdl-30289072

ABSTRACT

Antibody-toxin fused agents or immunotoxins, are a newly engineered class of cytotoxic agents consisting of a bacterial or plant toxin moiety hooked up either to a monoclonal antibody or a specific growth factor. Nevertheless, acquiring a full potency in clinic is mostly restricted due to the Capillary leak syndrome (CLS), a serious immune provoked, life-threatening side effect, subsequent to the endothelial damage, resulting in fluid escape from the bloodstream into tissues including lungs, muscle and brain, developing organ failure and eventually death. Proposed underlying mechanisms include direct damage to endothelial cells, acute inflammation, Lymphokine-activated killer (LAK) cells engagement, alteration in cell-cell/cell-matrix connectivities and cytoskeletal dysfunction. Very poor biodistribution and heterogeneous extravasation pattern in tumor site result in accumulation of ITs close to the extravasation site, gradual toxin release and initiation of nearby endothelial cells lysis, secretion of pro-inflammatory cytokines, development of acute inflammation and engagement of Lymphokine-activated killer (LAK) cells. Intrinsic immunogenicity of applied toxin moiety is another important determinant of CLS incidence. Toxins with more intrinsic immunogenicity possess more probability for CLS development. Recently, development of new generations of antibodies and mutated toxins with conserved cytotoxicity has partly tapered risk of CLS development. Here, we describe probable mechanisms involved in CLS and introduce some of the recently applied strategies for lessening incidence of CLS as much as possible.


Subject(s)
Capillary Leak Syndrome , Cytokines/immunology , Immunotoxins , Killer Cells, Lymphokine-Activated , Neoplasms , Animals , Capillary Leak Syndrome/chemically induced , Capillary Leak Syndrome/immunology , Capillary Leak Syndrome/pathology , Capillary Leak Syndrome/therapy , Humans , Immunotoxins/adverse effects , Immunotoxins/therapeutic use , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/pathology , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology
5.
Int J Mol Sci ; 19(9)2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30223493

ABSTRACT

HCV genotype 2a strain JFH-1 replicates and produces viral particles efficiently in human hepatocellular carcinoma (huh) 7.5 cells, which provide a stable in vitro cell infection system for the hepatitis C virus (HCVcc system). Natural killer (NK) cells are large lymphoid cells that recognize and kill virus-infected cells. In this study, we investigated the interaction between NK cells and the HCVcc system. IL-10 is a typical immune regulatory cytokine that is produced mostly by NK cells and macrophages. IL-21 is one of the main cytokines that stimulate the activation of NK cells. First, we used anti-IL-10 to neutralize IL-10 in a coculture of NK cells and HCVcc. Anti-IL-10 treatment increased the maturation of NK cells by enhancing the frequency of the CD56+dim population in NK-92 cells. However, with anti-IL-10 treatment of NK cells in coculture with J6/JFH-1-huh 7.5 cells, there was a significant decrease in the expression of STAT1 and STAT5 proteins in NK-92 cells and an increase in the HCV Core and NS3 proteins. In addition, rIL-21 treatment increased the frequency of the CD56+dim population in NK-92 cells, Also, there was a dramatic increase in the expression of STAT1 and STAT5 proteins in rIL-21 pre-stimulated NK cells and a decrease in the expression of HCV Core protein in coculture with J6/JFH-1-huh 7.5 cells. In summary, we found that the functional activation of NK cells can be modulated by anti-IL-10 or rIL-21, which controls the expression of HCV proteins as well as HCV RNA replication.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/virology , Host-Pathogen Interactions/immunology , Immunomodulation , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , CD56 Antigen/metabolism , Cell Line , Extracellular Signal-Regulated MAP Kinases , Humans , Interferon-gamma/metabolism , Interleukin-10/antagonists & inhibitors , Interleukin-10/metabolism , STAT Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Immunother ; 40(7): 265-276, 2017 09.
Article in English | MEDLINE | ID: mdl-28622272

ABSTRACT

Immune cell-derived exosomes can increase immunity against tumors. In contrast, tumor-derived exosomes can reduce the immunity and can change the tumor microenvironment to further develop and provide metastasis. These effects take place by an alteration in the innate and adaptive immune cell functions. In this experiment, we studied the natural killer (NK) cells' effectiveness on tumor cells after expansion and thereafter incubated it with exosomes. The exosomes were derived from 2 populations of NK cells: (1) naive NK cells and, (2) NK cells previously exposed to neuroblastoma (NB) cells. Moreover, we have studied the NB-derived exosomes on NK cell function. The molecular load of the characterized exosomes (by means of nanoparticle-tracking analysis, flow cytometry, scanning electron microscopy, and western blot) from NK cells exposed to the NB cell revealed their expression of natural killer cell receptors in addition to CD56, NKG2D, and KIR2DL2 receptors. These exosomes were used to treat NK cells and thereafter administered to NB tumor cells both in vitro and in vivo. Our results showed some kind of NK cells' education by the exosomes. This education from NK cells previously exposed to NB cell-derived exosomes caused efficient and greater cytotoxicity against NB tumors, but NB-derived exosomes act as tumor promoters by providing a tumor supporting niche. Hence, this method of preparing the exosomes has a dramatic effect on activation of anti-NK cells against NB cells.


Subject(s)
Cytokines/metabolism , Exosomes/metabolism , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neuroblastoma/immunology , Neuroblastoma/metabolism , Animals , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Heterografts , Humans , Immunophenotyping , Male , Mice , Neuroblastoma/mortality , Neuroblastoma/pathology , Phenotype , Receptors, Natural Killer Cell/metabolism , Tumor Burden
9.
J Pharmacol Exp Ther ; 361(1): 39-50, 2017 04.
Article in English | MEDLINE | ID: mdl-28154014

ABSTRACT

Dual specificity mitogen-activated protein kinase (MAPK) phosphatases [dual specificity phosphatase/MAP kinase phosphatase (DUSP-MKP)] have been hypothesized to maintain cancer cell survival by buffering excessive MAPK signaling caused by upstream activating oncogenic products. A large and diverse body of literature suggests that genetic depletion of DUSP-MKPs can reduce tumorigenicity, suggesting that hyperactivating MAPK signaling by DUSP-MKP inhibitors could be a novel strategy to selectively affect the transformed phenotype. Through in vivo structure-activity relationship studies in transgenic zebrafish we recently identified a hyperactivator of fibroblast growth factor signaling [(E)-2-benzylidene-5-bromo-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI-215)] that is devoid of developmental toxicity and restores defective MAPK activity caused by overexpression of DUSP1 and DUSP6 in mammalian cells. Here, we hypothesized that BCI-215 could selectively affect survival of transformed cells. In MDA-MB-231 human breast cancer cells, BCI-215 inhibited cell motility, caused apoptosis but not primary necrosis, and sensitized cells to lymphokine-activated killer cell activity. Mechanistically, BCI-215 induced rapid and sustained phosphorylation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) in the absence of reactive oxygen species, and its toxicity was partially rescued by inhibition of p38 but not JNK or ERK. BCI-215 also hyperactivated MKK4/SEK1, suggesting activation of stress responses. Kinase phosphorylation profiling documented BCI-215 selectively activated MAPKs and their downstream substrates, but not receptor tyrosine kinases, SRC family kinases, AKT, mTOR, or DNA damage pathways. Our findings support the hypothesis that BCI-215 causes selective cancer cell cytotoxicity in part through non-redox-mediated activation of MAPK signaling, and the findings also identify an intersection with immune cell killing that is worthy of further exploration.


Subject(s)
Breast Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/metabolism , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Female , HeLa Cells , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Killer Cells, Lymphokine-Activated/immunology , Mitogen-Activated Protein Kinase Phosphatases/immunology , Rats , Zebrafish
10.
Cell Physiol Biochem ; 41(2): 609-622, 2017.
Article in English | MEDLINE | ID: mdl-28214839

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignant diseases worldwide with limited therapeutic options. Accumulating evidences suggest that immunotherapy could be a promising option for treating HCC. T cell-associated immunotherapy lights up the hope for the improvement of complementary approach to conventional HCC treatments, which needs further research to consummate the clinical consequences. The present work reviewed several T cells associated cellular immunotherapies for HCC, including immune checkpoint blockade, gene-engineered T cells, bispecific T cell engagers, and so on. We also analyzed how these immunotherapies can mediate tumor cell eradication and evaluated their superiority or insufficiency.


Subject(s)
Carcinoma, Hepatocellular/therapy , Immunotherapy , Liver Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Carcinoma, Hepatocellular/pathology , Humans , Killer Cells, Lymphokine-Activated/cytology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Liver Neoplasms/pathology , Receptors, Antigen/genetics , Receptors, Antigen/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
11.
Vet Immunol Immunopathol ; 177: 58-63, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27436446

ABSTRACT

We evaluated changes in peripheral blood lymphocyte (PBL) count in dogs following adoptive immunotherapy using lymphokine-activated T killer cells (T-LAK) in combination with surgery. Fifteen tumor-bearing dogs treated with T-LAK therapy combined with palliative resection of tumors were enrolled in the present study. T-LAK were generated from autologous peripheral blood mononuclear cells (PBMC) by culture with recombinant human interleukin -2 (rhIL-2) and solid phase anti-canine cluster of differentiation (CD)3 antibody. T-LAK were administrated intravenously at 2-4-week intervals. After the first administration of T-LAK, counts of PBL and T lymphocyte subsets (CD3(+), CD4(+) and CD8(+) cells) increased and the CD4/CD8 ratio decreased, with significant increases in CD8(+) cells (P<0.05). In 8 tumor-bearing dogs that were administered sequential T-LAK, available data on changes in PBL and T lymphocyte phenotypes until the fifth administration were also analyzed. In tumor-bearing dogs administered 5 rounds of T-LAK, CD8(+) cell counts were maintained high until the fifth administration of T-LAK. Moreover, the CD4/CD8 ratio remained low until the fifth administration of T-LAK. These results indicate that T-LAK therapy combined with surgery may increase peripheral blood T lymphocytes, particularly CD8(+) cells, in tumor-bearing dogs.


Subject(s)
Immunotherapy, Adoptive/veterinary , Killer Cells, Lymphokine-Activated/immunology , Neoplasms/veterinary , Animals , Combined Modality Therapy/veterinary , Dogs , Female , Humans , Immunotherapy, Adoptive/methods , Lymphocyte Count , Male , Neoplasms/immunology , Neoplasms/therapy , Palliative Care , T-Lymphocyte Subsets/immunology
12.
Anticancer Res ; 36(5): 2307-13, 2016 May.
Article in English | MEDLINE | ID: mdl-27127137

ABSTRACT

BACKGROUND/AIM: Adoptive immunotherapy (AIT) using autologous zoledronate-activated killer (ZAK) cells has been performed for developing a novel modality of cancer treatment. In this study, data series from incurable pancreatic cancer were analyzed. PATIENTS AND METHODS: Patients were treated with AIT using intravenous administration of ZAK cells every 3 to 4 weeks in combination with standard chemotherapy and possible clinical benefits were examined. RESULTS: Seventy-five patients were treated. A median overall survival (OS) time of 6.7 months was achieved for all patients and 13.1 months for those treated 5 times or more, that increased to 14.6 and 18.3 months, respectively, when the previous treatment period of chemotherapy alone was included in the analysis. The disease control rate was 58.5 %. Multivariate regression analysis showed a significant positive correlation between the survival and baseline value of lymphocyte percentage in white blood cell counts (p=0.031). CONCLUSION: The data suggest that AIT using ZAK cells in combination with chemotherapy is safe and feasible and may be effective in prolonging survival for patients with incurable pancreatic cancer. The lymphocyte percentage at baseline may be a good biomarker for predicting the survival benefit of ZAK cell AIT.


Subject(s)
Diphosphonates/pharmacology , Imidazoles/pharmacology , Immunotherapy, Adoptive , Killer Cells, Lymphokine-Activated/immunology , Pancreatic Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cells, Cultured , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Combinations , Female , Humans , Interleukin-2/pharmacology , Kaplan-Meier Estimate , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/transplantation , Male , Middle Aged , Oxonic Acid/administration & dosage , Pancreatic Neoplasms/drug therapy , Prospective Studies , Salvage Therapy , Tegafur/administration & dosage , Treatment Outcome , Zoledronic Acid , Gemcitabine
13.
Sci Rep ; 6: 21678, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26902929

ABSTRACT

Merkel cell carcinoma (MCC) is a virally associated cancer characterized by its aggressive behavior and strong immunogenicity. Both viral infection and malignant transformation induce expression of MHC class I chain-related protein (MIC) A and B, which signal stress to cells of the immune system via Natural Killer group 2D (NKG2D) resulting in elimination of target cells. However, despite transformation and the continued presence of virally-encoded proteins, MICs are only expressed in a minority of MCC tumors in situ and are completely absent on MCC cell lines in vitro. This lack of MIC expression was due to epigenetic silencing via MIC promoter hypo-acetylation; indeed, MIC expression was re-induced by pharmacological inhibition of histone deacetylases (HDACs) both in vitro and in vivo. This re-induction of MICs rendered MCC cells more sensitive to immune-mediated lysis. Thus, epigenetic silencing of MICs is an important immune escape mechanism of MCCs.


Subject(s)
Carcinoma, Merkel Cell/genetics , Gene Silencing/immunology , Histocompatibility Antigens Class I/genetics , Histone Deacetylases/genetics , Killer Cells, Lymphokine-Activated/immunology , Skin Neoplasms/genetics , Acetylation/drug effects , Animals , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cytotoxicity, Immunologic , Gene Silencing/drug effects , Histocompatibility Antigens Class I/immunology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/immunology , Histones/genetics , Histones/immunology , Humans , Hydroxamic Acids/pharmacology , Killer Cells, Lymphokine-Activated/cytology , Killer Cells, Lymphokine-Activated/drug effects , Mice , Mice, Inbred NOD , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic/drug effects , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Vorinostat , Xenograft Model Antitumor Assays
14.
Microbiol Immunol ; 60(1): 47-55, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26616436

ABSTRACT

Performance status (PS) frequently improves occurs in cancer patients who have been infused with their own lymphokine-activated killer T cells (LAK-T). In the present study, a culture supernatant of LAK-T (LAK-T sup) administered to 8-week-old rats caused neurogenesis as evidenced by increased 5-ethynyl-2'-deoxyuridine staining of brain tissues. Intravenous injection of granulocyte-macrophage colony stimulating factor (GM-CSF), a major cytokine in LAK-T sup, had a similar effect. Furthermore, LAK-T sup induced Ca(++) increase in rat hippocampal brain slices that was detected in neuronal cells by emission of Fluo-8 NW at 520 nm. The same effect was observed with an rGM-CSF solution. GM-CSF may activate neuronal cells by stimulating the glial cells that surround and attach to them. If so, GM-CSF and LAK-T sup may improve the motor neurons of patients with amyotrophic lateral sclerosis. The neurogenerative effect of GM-CSF in LAK-T sup may also help improve brain function in aged adults including those with dementia such as Alzheimer's disease.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Neurons/immunology , T-Lymphocytes, Cytotoxic/drug effects , Adult , Alzheimer Disease/therapy , Amyotrophic Lateral Sclerosis/therapy , Animals , Brain/drug effects , Brain/immunology , Brain/pathology , Cell Proliferation/physiology , Cytokines/immunology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunotherapy, Adoptive/methods , Interleukin-2/immunology , Killer Cells, Lymphokine-Activated/cytology , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/transplantation , Male , Neurogenesis/immunology , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation
15.
Immunol Res ; 64(2): 548-57, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26659089

ABSTRACT

Follicular lymphoma (FL) is a disease of paradoxes-incurable but with a long natural history. We hypothesized that a combination of lymphokine-activated killer (LAK) cells and monoclonal antibodies might provide a robust synergistic treatment and tested this hypothesis in a phase II clinical trial (NCT01329354). In this trial, in addition to R-CHOP, we alternated the administration of only rituximab with rituximab and autologous LAK cells that were expanded ex vivo. Our objective was to determine the in vitro capability of LAK cells generated from FL patients to produce cytotoxicity against tumor cell lines and to determine rituximab- and obinutuzumab-induced cytotoxicity via antibody-dependent cellular cytotoxicity (ADCC) activity. We analyzed the LAK cell-induced cytotoxicity and rituximab (R)- and obinutuzumab (GA101)-induced ADCC activity. We show that LAK cells generated from FL patients induce cytotoxicity against tumor cell lines. R and GA101 enhance cytolysis through ADCC activity of LAK cells. Impaired LAK cell cytotoxicity and ADCC activity were detected in 50 % of patients. Percentage of NK cells in LAK infusions were correlated with the R- and GA101-induced ADCC. Our results indicate that the combination of R or GA101 and LAK cells should be an option as frontline maintenance therapy in patients with FL.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Lymphokine-Activated/immunology , Lymphoma, Follicular/immunology , Rituximab/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Killer Cells, Lymphokine-Activated/metabolism , Lymphocyte Count , Lymphoma, Follicular/diagnosis , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/metabolism , Phenotype , Rituximab/pharmacology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Time Factors
16.
J Exp Ther Oncol ; 11(3): 181-188, 2016 Jul.
Article in English | MEDLINE | ID: mdl-28471123

ABSTRACT

Pristimerin (PM) is a quinonemethide triterpenoid with cytotoxic activity against a wide range of cancer cell lines. However, the effect of PM on IL-2 induced activation of T lymphocytes, which play a major role in antitumor immunity has not been studied. The objective of the present study was to evaluate the effect of PM on IL-2 induced proliferation of T cells, generation of lymphokine activated killer cells (LAK cells) and the signaling pathways involved in activation of T cells by IL-2. PM inhibited the IL-2 induced proliferation of mouse splenic T cells and the generation LAK cells at very low concentrations. The suppression of T cell proliferation by PM was associated with the inhibition of IL-2 induced Janus kinase/signal transducers and activators of transcription (Jak/STAT) and extracellular signal-regulated kinase 1 and 2 (Erk1/2) signaling pathways. PM also inhibited the proliferation and differentiation-related immediate early gene products such as p-c-fos, p-c-jun, c-myc and cyclin D1. In addition, antiapoptotic (prosurvival) NF-кB, p-Akt and p-mTOR were also inhibited by PM. These data demonstrated that PM inhibits IL-2 induced T cell activation and generation of LAK cells by disrupting multiple cell signaling pathways induced by IL-2.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Spleen/drug effects , T-Lymphocytes/drug effects , Triterpenes/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Janus Kinases/metabolism , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Mice , Pentacyclic Triterpenes , Phosphorylation , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Oncotarget ; 6(36): 39342-56, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26513172

ABSTRACT

The antitumorigenic mechanism of the selective cyclooxygenase-2 (COX-2) inhibitor celecoxib is still a matter of debate. Using lung cancer cell lines (A549, H460) and metastatic cells derived from a lung cancer patient, the present study investigates the impact of celecoxib on the expression of intercellular adhesion molecule 1 (ICAM-1) and cancer cell lysis by lymphokine-activated killer (LAK) cells. Celecoxib, but not other structurally related selective COX-2 inhibitors (i.e., etoricoxib, rofecoxib, valdecoxib), was found to cause a substantial upregulation of ICAM-1 protein levels. Likewise, ICAM-1 mRNA expression was increased by celecoxib. Celecoxib enhanced the susceptibility of cancer cells to be lysed by LAK cells with the respective effect being reversed by a neutralizing ICAM-1 antibody. In addition, enhanced killing of celecoxib-treated cancer cells was reversed by preincubation of LAK cells with an antibody to lymphocyte function associated antigen 1 (LFA-1), suggesting intercellular ICAM-1/LFA-1 crosslink as crucial event within this process. Finally, celecoxib elicited no significant increase of LAK cell-mediated lysis of non-tumor bronchial epithelial cells, BEAS-2B, associated with a far less ICAM-1 induction as compared to cancer cells. Altogether, our data demonstrate celecoxib-induced upregulation of ICAM-1 on lung cancer cells to be responsible for intercellular ICAM-1/LFA-1 crosslink that confers increased cancer cell lysis by LAK cells. These findings provide proof for a novel antitumorigenic mechanism of celecoxib.


Subject(s)
Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Killer Cells, Lymphokine-Activated/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Cell Line, Tumor , Humans , Intercellular Adhesion Molecule-1/immunology , Killer Cells, Lymphokine-Activated/immunology , Lung Neoplasms/immunology , Transfection , Up-Regulation/drug effects
18.
Cancer Lett ; 368(1): 54-63, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26276724

ABSTRACT

Current therapies fail to cure most metastatic or recurrent bone cancer. We explored the efficacy and the pathways involved in natural killer (NK) cells' elimination of osteosarcoma (OS) cells, including tumor initiating cells (TICs), which are responsible for chemotherapy resistance, recurrence, and metastasis. The expression of ligands for NK cell receptors was studied in primary OS cell lines by flow cytometry. In vitro cytotoxicity of activated and expanded NK (NKAE) cells against OS was tested, and the pathways involved explored by using specific antibody blockade. NKAE cells' ability to target OS TICs was analyzed by flow cytometry and sphere formation assays. Spironolactone (SPIR) was tested for its ability to increase OS cells' susceptibility to NK cell lysis in vitro and in vivo. We found OS cells were susceptible to NKAE cells' lysis both in vivo and in vitro, and this cytolytic activity relied on interaction between NKG2D receptor and NKG2D ligands (NKG2DL). SPIR increased OS cells' susceptibility to lysis by NKAE cells, and could shrink the OS TICs. Our results show NKAE cells target OS cells including the TICs compartment, supporting the use of NK-cell based immunotherapies for OS.


Subject(s)
Bone Neoplasms/therapy , Cell Proliferation , Immunotherapy, Adoptive/methods , Killer Cells, Lymphokine-Activated/transplantation , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplastic Stem Cells/metabolism , Osteosarcoma/therapy , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/immunology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Communication , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Humans , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/metabolism , Ligands , Mice, Inbred NOD , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Osteosarcoma/immunology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Signal Transduction , Spironolactone/pharmacology , Time Factors
19.
Oncotarget ; 6(2): 1079-89, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25473902

ABSTRACT

The patient-derived tumor xenograft (PDTX) models can reproduce a similar natural genetic background and similar biological behaviors to tumor cells in patients, which is conducive to the assessment of personalized cancer treatment. In this study, to verify the targeting and effectiveness of the therapeutic strategy using a Survivin promoter-regulated oncolytic adenovirus expressing Hsp70, the PDTX models of hepatocellular carcinoma (HCC) were established in nude mice and the cytokine-induced killer (CIK) cells were intravenously infused into mice to partially reconstruct the mouse immune function. The results demonstrated that, either the immune anti-tumor effect caused by CIK cell infusion or the oncolytic effect generated by oncolytic adenovirus replication was very limited. However, the synergistic tumor inhibitory effect was significantly enhanced after treatments with oncolytic adenovirus expressing Hsp70 combined with CIK cells. Oncolytic adenovirus mediated the specific expression of Hsp70 in cancer tissues allowed the CIK chemotaxis, and induce the infiltration of CD3+ T cells in tumor stroma, thereby exhibiting anti-tumor activity. The anti-tumor effect was more effective for the highly malignant tumor xenografts with highly Survivin expression. This strategy can synergistically activate multiple anti-tumor mechanisms and exert effective anti-tumor activities that have a significant inhibitory effect against the growth of HCC xenografts.


Subject(s)
Carcinoma, Hepatocellular/therapy , Genetic Therapy/methods , HSP70 Heat-Shock Proteins/metabolism , Immunotherapy, Adoptive/methods , Liver Neoplasms/therapy , Oncolytic Virotherapy/methods , Xenograft Model Antitumor Assays/methods , Adenoviridae/genetics , Adult , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Combined Modality Therapy , Gene Expression , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/transplantation , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Oncolytic Viruses/genetics , Survivin , Treatment Outcome , Tumor Burden
20.
J Cancer Res Clin Oncol ; 141(7): 1249-58, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25381064

ABSTRACT

PURPOSE: The aim of the present study was to discuss recent findings on the role of T cells in lung cancer to provide information on their potential application, especially in cellular immunotherapy. METHODS: Data on the different types of T cells that are currently used for the treatment of lung cancer were obtained by searching the PUBMED database. RESULTS: Cytotoxic T lymphocytes, natural killer T cells, γδ T cells, lymphokine-activated killer cells, tumor-infiltrating lymphocytes, cytokine-induced killer cells and gene-modified T cells were analyzed to determine the benefits and drawbacks of their application in the treatment of lung cancer. Advances in the study of their antitumor mechanisms and directions for future research were discussed. CONCLUSIONS: T cells are critical for tumorigenesis and therefore important targets for the treatment of lung cancer. T-cell-associated cellular immunotherapy opens up a window of opportunity for the development of complementary methods to traditional lung cancer treatments, which warrants further investigation to improve the clinical outcomes of lung cancer patients.


Subject(s)
Immunotherapy, Adoptive/methods , Lung Neoplasms/therapy , T-Lymphocytes/transplantation , Cytokine-Induced Killer Cells/immunology , Cytokine-Induced Killer Cells/transplantation , Cytotoxicity, Immunologic/physiology , Humans , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/transplantation , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...