Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
J Immunother Cancer ; 11(8)2023 08.
Article in English | MEDLINE | ID: mdl-37536936

ABSTRACT

BACKGROUND: The tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine is made by ex vivo priming matured autologous dendritic cells (DCs) with yeast cell wall particles (YCWPs) loaded with autologous tumor lysate (TL). The tumor lysate, particle only (TLPO) vaccine uses autologous TL-loaded YCWPs coated with silicate for in vivo DC loading. Here we report the 36-month prespecified analyses of this prospective, randomized, double-blind trial investigating the ability of the TLPO and TLPLDC (±granulocyte-colony stimulating factor (G-CSF)) vaccines to prevent melanoma recurrence in high-risk patients. METHODS: Patients with clinically disease-free stage III/IV melanoma were randomized 2:1 initially to TLPLDC versus placebo (n=124) and subsequently TLPO versus TLPLDC (n=63). All patients were randomized and blinded; however, the placebo control arm was replaced in the second randomization scheme with another novel vaccine; some analyses in this paper therefore reflect a combination of the two randomization schemes. Patients receiving the TLPLDC vaccine were further divided by their method of DC harvest (with or without G-CSF pretreatment); this was not randomized. The use of standard of care checkpoint inhibitors was not stratified between groups. Safety was assessed and Kaplan-Meier and log-rank analyses compared disease-free (DFS) and overall survival (OS). RESULTS: After combining the two randomization processes, a total of 187 patients were allocated between treatment arms: placebo (n=41), TLPLDC (n=103), or TLPO (n=43). The allocation among arms created by the addition of patients from the two separate randomization schemes does not reflect concurrent randomization among all treatment arms. TLPLDC was further divided by use of G-CSF in DC harvest: no G-CSF (TLPLDC) (n=47) and with G-CSF (TLPLDC+G) (n=56). Median follow-up was 35.8 months. Only two patients experienced a related adverse event ≥grade 3, one each in the TLPLDC+G and placebo arms. DFS was 27.2% (placebo), 55.4% (TLPLDC), 22.9% (TLPLDC+G), and 60.9% (TLPO) (p<0.001). OS was 62.5% (placebo), 93.6% (TLPLDC), 57.7% (TLPLDC+G), and 94.6% (TLPO) (p=0.002). CONCLUSIONS: The TLPO and TLPLDC (without G-CSF) vaccines were associated with improved DFS and OS in this clinical trial. Given production and manufacturing advantages, the efficacy of the TLPO vaccine will be confirmed in a phase 3 trial. TRIAL REGISTRATION NUMBER: NCT02301611.


Subject(s)
Cancer Vaccines , Melanoma , Humans , Prospective Studies , Cancer Vaccines/therapeutic use , Dendritic Cells , Granulocyte Colony-Stimulating Factor , Melanoma, Cutaneous Malignant
2.
Cancer Immunol Immunother ; 72(3): 697-705, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36045304

ABSTRACT

BACKGROUND: A randomized, double-blind, placebo-controlled phase 2b trial of the tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine was conducted in patients with resected stage III/IV melanoma. Dendritic cells (DCs) were harvested with and without granulocyte-colony stimulating factor (G-CSF). This analysis investigates differences in clinical outcomes and RNA gene expression between DC harvest methods. METHODS: The TLPLDC vaccine is created by loading autologous tumor lysate into yeast cell wall particles (YCWPs) and exposing them to phagocytosis by DCs. For DC harvest, patients had a direct blood draw or were pretreated with G-CSF before blood draw. Patients were randomized 2:1 to receive TLPLDC or placebo. Differences in disease-free survival (DFS) and overall survival (OS) were evaluated. RNA-seq analysis was performed on the total RNA of TLPLDC + G and TLPLDC vaccines to compare gene expression between groups. RESULTS: 144 patients were randomized: 103 TLPLDC (47 TLPLDC/56 TLPLDC + G) and 41 placebo (19 placebo/22 placebo + G). Median follow-up was 27.0 months. Both 36-month DFS (55.8% vs. 24.4% vs. 30.0%, p = 0.010) and OS (94.2% vs. 69.8% vs. 70.9%, p = 0.024) were improved in TLPLDC compared to TLPLDC + G or placebo, respectively. When compared to TLPLDC + G vaccine, RNA-seq from TLPLDC vaccine showed upregulation of genes associated with DC maturation and downregulation of genes associated with DC suppression or immaturity. CONCLUSIONS: Patients receiving TLPLDC vaccine without G-CSF had improved OS and DFS. Outcomes remained similar between patients receiving TLPLDC + G and placebo. Direct DC harvest without G-CSF had higher expression of genes linked to DC maturation, likely improving clinical efficacy.


Subject(s)
Cancer Vaccines , Melanoma , Humans , Dendritic Cells , Granulocyte Colony-Stimulating Factor , Melanoma, Cutaneous Malignant
3.
Vaccine ; 36(23): 3247-3253, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29724512

ABSTRACT

INTRODUCTION: Tumor vaccines use various strategies to generate immune responses, commonly targeting generic tumor-associated antigens. The tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine is produced from DC loaded with autologous tumor antigens, creating a patient-specific vaccine. Here, we describe initial phase I/IIa trial results. METHODS: This trial includes patients with any stage solid tumors, ECOG ≤1, and >4 months life-expectancy. A personalized vaccine is created using 1 mg of tumor and 120 ml blood (to isolate DC). Primary vaccination series (PVS) is four monthly inoculations. Patients are followed per standard of care (SOC). Endpoints include safety and tumor response (RECIST v1.1). RESULTS: 44 patients were enrolled and vaccinated consisting of 31 late stage patients with residual/measurable disease, and 13 disease-free patients after SOC therapies. While 4 patients progressed before completing the PVS, 12/31 (39%) demonstrated clinical benefit (2 complete responses, 4 partial responses, 6 stable disease). In the adjuvant setting, 46% of late stage patients remain disease free at a median of 22.5 months. CONCLUSIONS: The TLPLDC vaccine is scalable, generates a personalized DC vaccine, and requires little autologous tumor tissue and few DC. The vaccine is safe, with primarily grade 0-2 toxicities, and nearly 40% clinical benefit rate in varied tumors, warranting further study. TRIAL REGISTRATION: ISRCTN81339386, Registered 2/17/2016.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/therapy , Adult , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Female , Humans , Male , Middle Aged , Neoplasms/pathology , Precision Medicine/methods , Treatment Outcome
4.
Immunotherapy ; 10(5): 373-382, 2018 04.
Article in English | MEDLINE | ID: mdl-29473470

ABSTRACT

AIM: We developed a novel approach to efficiently deliver autologous tumor antigens to the cytoplasm of dendritic cells (DC) using yeast cell wall particles (YCWP). MATERIALS AND METHODS: Loading of YCWP, leakage of protein from loaded YCWP and cytoplasmic delivery of YCWP content was assessed using fluorescent-tagged experiments. Spectrophotometric analysis compared the epitope-specific T-cell responses following antigen presentation via YCWP versus exogenous loading. The in vivo effectiveness of tumor lysate (TL) particle loaded DC (TLPLDC) vaccine was assessed using murine melanoma models. RESULTS: In fluorescence-tagged experiments, YCWP efficiently delivered antigen to the cytoplasm of DC. TLPLDC loading was more effective than conventional exogenous loading of DC. Finally, in murine melanoma models, TLPLDC outperformed an analogous dendritoma vaccine. CONCLUSION: The TLPLDC vaccine is commercially scalable and holds the potential of producing personalized vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Disease Models, Animal , Female , Immunotherapy/methods , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL
5.
Cancer Immunol Immunother ; 65(4): 383-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26894495

ABSTRACT

BACKGROUND: Stage IV melanoma has high mortality, largely unaffected by traditional therapies. Immunotherapy including cytokine therapies and checkpoint inhibitors improves outcomes, but has significant toxicities. In this phase I/IIa trial, we investigated safety and efficacy of a dendritoma vaccine, an active, specific immunotherapy, in stage IV melanoma patients. METHODS: Autologous tumor lysate and dendritic cells were fused creating dendritoma vaccines for each patient. Phase I patients were vaccinated every 3 months with IL-2 given for 5 days after initial inoculation. Phase IIa patients were vaccinated every 6 weeks with IL-2 given on days 1, 3 and 5 after initial inoculation. Toxicity and clinical outcomes were assessed. RESULTS: Twenty-five patients were enrolled and inoculated. All dendritoma and IL-2 toxicities were

Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Interleukin-2/immunology , Melanoma/immunology , Melanoma/therapy , Aged , Arthralgia/chemically induced , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Chills/chemically induced , Combined Modality Therapy , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Erythema/chemically induced , Female , Humans , Interleukin-2/adverse effects , Interleukin-2/therapeutic use , Kaplan-Meier Estimate , Male , Middle Aged , Nausea/chemically induced , Neoplasm Recurrence, Local , Neoplasm Staging , Treatment Outcome
6.
Oncol Lett ; 8(1): 198-202, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24959245

ABSTRACT

Although the generation of hybrid cells by cell fusion plays a significant role in biotechnology and biomedicine, the low cell-fusion rates and the limitation of large-scale cell fusion for clinical applications of the two widely used approaches, polyethylene-glycol (PEG)-mediated cell fusion and electrofusion, hinder the application of this critical technology in certain key areas, including cancer immunotherapy. In the present study, a simple procedure that can not only significantly increase the heterologous cell fusion but is also capable of producing fused cells on a large scale is reported. A biotin-streptavidin-biotin (BSB) bridge was created by coating one to-be-fused cell with biotin and the other with biotin-streptavidin. The BSB bridge enhances cell-fusion rates induced with PEG fusion or electrofusion by 10-30% depending on the cell types when compared with cell fusions without the bridge. The procedure described increases heterologous cell pairing and eliminates the alignment step required for the majority of electrofusions. Notably, it can be used to make large-scale cell fusions for clinical applications.

7.
Aging Cell ; 13(1): 102-10, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23957304

ABSTRACT

Disruption of adenylyl cyclase type 5 (AC5) knockout (KO) is a novel model for longevity. Because malignancy is a major cause of death and reduced lifespan in mice, the goal of this investigation was to examine the role of AC5KO in protecting against cancer. There have been numerous discoveries in genetically engineered mice over the past several decades, but few have been translated to the bedside. One major reason is that it is difficult to alter a gene in patients, but rather a pharmacological approach is more appropriate. The current investigation employs a parallel construction to examine the extent to which inhibiting AC5, either in a genetic knockout (KO) or by a specific pharmacological inhibitor protects against cancer. This study is unique, not only because a combined genetic and pharmacological approach is rare, but also there are no prior studies on the extent to which AC5 affects cancer. We found that AC5KO delayed age-related tumor incidence significantly, as well as protecting against mammary tumor development in AC5KO × MMTV-HER-2 neu mice, and B16F10 melanoma tumor growth, which can explain why AC5KO is a model of longevity. In addition, a Food and Drug Administration approved antiviral agent, adenine 9-ß-D-arabinofuranoside (Vidarabine or AraAde), which specifically inhibits AC5, reduces LP07 lung and B16F10 melanoma tumor growth in syngeneic mice. Thus, inhibition of AC5 is a previously unreported mechanism for prevention of cancers associated with aging and that can be targeted by an available pharmacologic inhibitor, with potential consequent extension of lifespan.


Subject(s)
Adenylyl Cyclases/genetics , Gene Deletion , Longevity , Melanoma/enzymology , Melanoma/pathology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Female , Longevity/drug effects , Mice , Mice, Knockout
8.
Int J Oncol ; 41(2): 661-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22581115

ABSTRACT

The dependency of activated natural killer (NK) cells on the continuous support of exogenous interleukin (IL)-2 for their in vivo survival, tumor localization and consequently, their antitumor effect, is a major obstacle for NK cell-mediated tumor therapy. In the present study, a fusion gene between IL-12 and mouse sonic hedgehog C-terminal domain (Shh-C) was constructed. The fusion protein was autocatalytically processed to form cholesterol-modified IL-12 molecules and an autocrine loop of IL-12 was established for the sustained activation of NK cells. The transduced NK cells matured more rapidly in vitro with the enhanced expression of granule-related proteins. NKIL-12/Shh-C cells reached the same proliferation rate as NK cells transduced with enhanced green fluorescent protein (EGFP)/Shh-C (NKEGFP/Shh-C) with <10-fold IL-2 supplementation, suggesting that the fusion protein reduced the dependency of NK cells on IL-2. The amount of interferon­Î³ (IFN-γ) in the supernatants of NKIL-12/Shh-C cells 5 and 7 days after transduction was significantly higher than that in the supernatants of NKIL-12 cells. Immunofluorescent staining of lung tissues from B16-bearing mice which had received an intravenous injection of lentivirus-transduced NK cells without exogenous IL-2 confirmed that donor NK cells successfully infiltrated into the lung tissues. The survival time of the mice which had received NKIL-12/Shh-C cells was significantly prolonged compared to the mice which had received NKEGFP/Shh-C cells.


Subject(s)
Autocrine Communication , Hedgehog Proteins/genetics , Interleukin-12/genetics , Killer Cells, Natural/physiology , Lymphocyte Activation , Melanoma, Experimental/therapy , Recombinant Fusion Proteins/genetics , Adoptive Transfer , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cytokines/blood , Cytokines/metabolism , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic , Female , Hedgehog Proteins/biosynthesis , Humans , Immunotherapy , Interleukin-12/biosynthesis , Interleukin-2/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Lung/pathology , Melanoma, Experimental/blood , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Transduction, Genetic
9.
Mol Med Rep ; 5(3): 788-92, 2012 03.
Article in English | MEDLINE | ID: mdl-22160152

ABSTRACT

The Akt pathway is considered a pivotal player in regulating cell survival, growth, migration and angiogenesis. Disruption of normal Akt/PKB/PTEN signaling frequently occurs in numerous types of human cancers. Therefore, this signaling pathway is regarded as an important target for effective cancer therapeutic strategies. In the present study, methanol extracts from Scutellaria barbata (S. barbata) were determined to be Akt/protein kinase B inhibitory, after screening a panel of 40 traditional Chinese herbs with the Fast Activated Cell-based ELISA (FACE) assay. S. barbata extracts were found to suppress the phosphorylation levels of Akt. This inhibition was Akt kinase-specific as it had no effect on PI3K, the upstream kinase of Akt, whereas the levels of phosphorylated Bad and FHKR, the two downstream targets of Akt, changed as the levels of Akt changed. S. barbata extracts also exhibited cytotoxicity against LoVo and human umbilical vein endothelial cells (HUVECs). Furthermore, this extract inhibited the process of in vitro angiogenesis of HUVECs on Matrigel. S. barbata may be a suitable alternative source with which to isolate small molecules for use as Akt kinase inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Scutellaria/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/toxicity , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/toxicity , Cell Line , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phosphorylation/drug effects , Plant Extracts/chemistry , Plant Extracts/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
10.
Am J Physiol Heart Circ Physiol ; 299(3): H707-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562336

ABSTRACT

Adenylyl cyclase (AC) type 5 (AC5) and AC type 6 (AC6) are the two major AC isoforms in the heart. Cardiac overexpression of AC6 has been shown to be protective in response to several interventions. In this investigation, we examined the effects of chronic pressure overload in AC6 transgenic (TG) mice. In the absence of any stress, AC6 TG mice exhibited enhanced contractile function compared with their wild-type (WT) littermates, i.e., increased (P < 0.05) left ventricular (LV) ejection fraction (EF) (75 +/- 0.9 vs. 71 +/- 0.5%) and LV dP/dt (7,850 +/- 526 vs. 6,374 +/- 315 mmHg/s). Forskolin (25 microg x kg(-1) x min(-1) for 5 min) increased LVEF more (P < 0.05) in AC6 TG mice (14.8 +/- 1.0%) than in WT mice (7.7 +/- 1.0%). Also, isoproterenol (0.04 microg x kg(-1) x min(-1) for 5 min) increased LVEF more (P < 0.05) in AC6 TG mice (18.0 +/- 1.2%) than in WT mice (11.6 +/- 2.1%). Pressure overload, induced by 4 wk of transverse aortic constriction (TAC), increased the LV weight-to-body weight ratio and myocyte cross-sectional area similarly in both groups, but reduced LVEF more in AC6 TG mice (22%) compared with WT mice (9%), despite the higher starting level of LVEF in AC6 TG mice. LV systolic wall stress increased more in AC6 TG mice than in WT mice, which could be responsible for the reduced LVEF in AC6 TG mice with chronic pressure overload. In addition, LV dP/dt was no longer elevated in AC6 TG mice after TAC compared with WT mice. LV end-diastolic diameter was also greater (P < 0.05) in AC6 TG mice (3.8 +/- 0.07 mm) than in WT mice (3.6 +/- 0.05 mm) after TAC. Thus, in contrast to other interventions previously reported to be salutary with cardiac AC6 overpression, the response to chronic pressure overload was not; actually, AC6 TG mice fared worse than WT mice. The mechanism may be due to the increased LV systolic wall stress in AC6 TG mice with chronic pressure overload.


Subject(s)
Adenylyl Cyclases/metabolism , Heart Ventricles/metabolism , Heart/physiopathology , Hypertrophy, Left Ventricular/metabolism , Myocardium/metabolism , Ventricular Dysfunction, Left/metabolism , Adenylyl Cyclases/genetics , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/physiology , Colforsin/pharmacology , Echocardiography , Heart/drug effects , Heart Rate/drug effects , Heart Rate/physiology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics , Hypertrophy, Left Ventricular/physiopathology , Isoproterenol , Mice , Mice, Transgenic , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Stress, Physiological/physiology , Ventricular Dysfunction, Left/physiopathology
11.
Oncol Rep ; 23(2): 545-50, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20043120

ABSTRACT

Dendritic cell-mediated cancer immunotherapy employs several ways to engage tumor antigens. We have demonstrated in both pre-clinical animal studies and early clinical trials that dendritomas, highly purified hybrids between dendritic cells (DC) and tumor cells, are superior activators of anti-tumor immunity. It has been argued, however, that DC vaccines may be dysfunctional in lymph node migration. In the present study we examined inflammatory chemokine and chemokine receptor expression as well as other maturation induced genes in dendritomas produced from either immature or mature DCs in order to shed light on their capacity to migrate from injection sites to draining lymph nodes and elicit an appropriate immune response. RNA microarray analysis was used to identify gene expression profiles for inflammatory chemokines and receptors and other maturation induced genes within dendritomas, lysate-pulsed dendritic cells, immature DCs and mature DCs. Gene regulation was confirmed with relative quantification, real-time RT-PCR in a separate experiment. We found that fusion of immature DCs to tumor cells initiates maturation with respect to inflammatory chemokines, chemokine receptors and other maturation induced genes in a similar pattern as LPS matured DCs. Interestingly, we saw a reversed gene profile when mature DCs were fused to tumor cells. LPS matured DCs displayed the chemokine repertoire expected with DC maturation; however, once fused to tumor cells, these chemokines and other maturation induced genes reverted to levels comparable to immature DCs. It appears that mature DCs used for dendritoma production result in a de-mature genotype. Our results indicate that dendritomas from immature DC/tumor cell fusions may be more effective in migration from injection site to draining lymph nodes and, therefore, would be more effective in stimulating anti-tumor immunity.


Subject(s)
Cell Dedifferentiation/physiology , Dendritic Cells/pathology , Dendritic Cells/physiology , Hybridomas/metabolism , Inflammation Mediators/metabolism , Animals , Cell Dedifferentiation/drug effects , Cell Differentiation/drug effects , Cell Extracts/pharmacology , Cell Fusion , Cell Movement/physiology , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Gene Expression Profiling , Hybridomas/immunology , Hybridomas/pathology , Immunity, Cellular/physiology , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
12.
Int J Oncol ; 35(4): 829-35, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19724919

ABSTRACT

Chemotherapy is one of the main treatment options for cancer, but the effectiveness of chemotherapeutic drugs is severely limited due to their systemic toxicity. Therefore, the need for a more targeted approach in tumor treatment is obvious. A tumor-activated agent would decrease systemic toxicity as well as increase the efficacy of the treatment. It has previously been shown that the latency of pro-TGF-beta is conferred by dimerization of two latency-associated peptides (LAP) that form a protective shield, which is cleaved off upon activation by matrix metalloproteinases (MMPs). It has also been shown that the fusion of this LAP peptide with other cytokines can confer their latency. In the present study, a recombinant adenovirus with a fusion gene encoding a tumor-activated pro-cytolytic peptide was made in which the LAP domain of TGF-beta was fused with melittin, a potent cytolytic toxin, with an MMP2 cleavage site in between the two. In vitro studies show that the melittin-MMP2-LAP recombinant adenovirus can be activated by MMP2 which leads to the release of free melittin to lyse the target cells. In vivo studies show approximately a 70% decrease in B16 tumor volume in melittin-MMP2-LAP recombinant adenovirus-treated mice as compared to control mice. No significant systemic toxicity was observed in the treated mice.


Subject(s)
Genetic Therapy , Latent TGF-beta Binding Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Melanoma, Experimental/therapy , Melitten/metabolism , Transforming Growth Factor beta/metabolism , Adenoviridae/genetics , Animals , CHO Cells , Cell Survival , Cricetinae , Cricetulus , Female , Genetic Vectors , Latent TGF-beta Binding Proteins/biosynthesis , Latent TGF-beta Binding Proteins/genetics , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Melitten/biosynthesis , Melitten/genetics , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
13.
Oncol Rep ; 21(5): 1147-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19360287

ABSTRACT

Noni juice as a folk medicine has been used for over two thousand years. Recently, some active ingredients of Noni juice have been successfully isolated and intensively studied. Because dendritic cells (DCs) are central regulators both in priming innate and adaptive immune responses and in maintaining self tolerance, in the current study we treated DCs with fermented Noni Exudate (fNE) in order to explore their function in regulating other immune cells. It was shown that fNE-treated DCs stimulate proliferation of splenocytes, among which, B cells are the major responsive cell group. The proliferative response of B cells to fNE-treated DCs is cell contact-dependent, CD40L-independent; and the adhesion feature of DCs was enhanced to form large DC-B conjugation cluster. Moreover, it was demonstrated that fNE-treated DCs promote B cell differentiation and Ig class switching. These results lay a foundation for the further exploration of fNE as a biological response modifier in the immune system.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Morinda/chemistry , Plant Extracts/pharmacology , Animals , CD40 Ligand/immunology , CD40 Ligand/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Growth Processes/drug effects , Cell Growth Processes/immunology , Cells, Cultured , Female , Fermentation , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL
14.
Vaccine ; 27(4): 558-64, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19027812

ABSTRACT

Whole tumor cell vaccines have been widely studied and remain promising cancer immunotherapies. In the present study, we discovered that vaccination with irradiated mouse sarcoma S180 tumor cells stimulated robust antitumor immunity to autologous tumor cells in both syngenic and allogenic mice. The antitumor activity requires both T and B cells, but not NK cells. When a mouse lung carcinoma (TC-1) whole tumor cell vaccine was combined with the S180 vaccine, the antitumor immunity against live TC-1 tumor cells is significantly enhanced compared to a TC-1 whole cell vaccine alone. This antitumor immunity not only prevents live tumor challenge but also eradicates existing tumor cells. A similar phenomenon was also observed when S180 vaccine was combined with LL2 Lewis lung carcinoma tumor cells. Therefore, S180 vaccine may serve as an adjuvant for other whole tumor cell vaccines.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor/radiation effects , Immunotherapy/methods , Sarcoma 180/immunology , Adjuvants, Immunologic , Animals , Cancer Vaccines/genetics , Female , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/immunology , Sarcoma 180/genetics , Vaccines, Synthetic/immunology
15.
Mol Med Rep ; 2(3): 399-403, 2009.
Article in English | MEDLINE | ID: mdl-21475841

ABSTRACT

We previously designed a pro-cytolytic peptide to target prostate-specific membrane antigen (PSMA)-positive prostate tumor cells. The backbone of the peptide was derived from the cell lytic amoebapore H-3 domain, which becomes completely inactive upon modification by two glutamate residues linked to the ε-amide group of the COOH-terminal lysine through γ-linkages (H-3Glu2). This modified H-3 domain regains its lytic activity against PSMA-positive cells (LNCaP) after the γ-linked glutamate residues are cleaved by PSMA. Our previous in vitro results demonstrate that the modified amoebapore peptide has strong cytolytic activity towards PSMA-positive cells and very little activity towards PSMA-negative cells. In the present study, the in vivo efficacy of this modified peptide was examined in human LNCaP prostate tumor xenografts in nude mice. The results showed significantly decreased tumor size and PSA levels in treated mice as compared to control mice. As well, 5/12 of the treated mice were tumor-free. Peptide distribution studies showed that peptide levels in the prostate tumors maintained a steady concentration for approximately 6 hours. Single-dose toxicity studies showed no toxic effects of the peptide when administered intraperitoneally or intravenously at a dose of 30 mg/kg.

16.
Oncol Rep ; 20(6): 1505-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020734

ABSTRACT

The anti-tumor activity of Morinda citrifolia fruit juice (Noni) has been previously reported. However, the mechanism behind this activity remains unknown. In the present study, we studied the anti-tumor activity of fermented Noni exudate (fNE) and demonstrated that intraperitoneal injection of this material significantly increased the percentages of granulocytes and NK cells in the peripheral blood, peritoneum, and spleen. Furthermore, in preventive and treatment settings, fNE injection induced complete tumor rejection in normal C57BL/6J mice, partial tumor rejection in C57 nude mice lacking functional lymphocytes, and no tumor rejection in NK cell deficient beige mice. Over 85% of the C57BL/6J mice that received fNE survived the first tumor injection and rejected up to 5 x 10(6) tumor cells when re-challenged. The anti-tumor activity remains in the heat-inactivated and filtrated supernatant of fNE. These data demonstrate that fNE appears to be able to stimulate the innate immune system and the adaptive immune system to reject tumor cells. NK cells respond quickly and appear to be among the major players of the innate immune system, while the adaptive immune system reacts later with a retained memory.


Subject(s)
Antineoplastic Agents/pharmacology , Exudates and Transudates/metabolism , Morinda/metabolism , Plant Extracts/metabolism , Animals , Ascites/metabolism , Female , Fermentation , Fruit/metabolism , Immune System , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Sarcoma/drug therapy
17.
Oncol Rep ; 18(3): 665-71, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17671717

ABSTRACT

Vaccination using dendritic/tumor cell hybrids represents a novel and promising cancer immunotherapy. We have developed a technology that can instantly purify the hybrids (dendritomas) from the fusion mixture of dendritic cells (DCs) and tumor cells. Our animal studies and a phase I study of stage IV melanoma patients demonstrated that dendritoma vaccination could be conducted without major toxicity and induced tumor cell-specific immunological and clinical responses. In this pilot study, ten stage IV renal cell carcinoma patients were studied. Dendritomas were made from autologous DCs and tumor cells and administered by subcutaneous injection. After initial vaccination, three escalating doses of IL-2 (3, 6, and 9 million units each) were followed within five days. This treatment regimen was tolerated well without severe adverse events directly related to the dendritoma vaccine. Most adverse events were related to IL-2 administration or pre-existing disease. Patient-specific immune responses were evaluated by flow cytometric measurement of interferon-gamma-producing T-cells before and after vaccination in response to stimulation with tumor antigens. Nine out of nine patients eligible for the analysis showed an increase of IFN-gamma-expressing CD4+ T cells after vaccination(s); while five out of eight patients eligible for the analysis showed an increase of IFN-gamma-expressing CD8+ T cells. Clinical responses were documented in 40% of the patients, three with stabilization of disease and one with a partial response documented by a reduction in tumor size. This pilot study demonstrated that dendritoma vaccines could be administered safely to patients with metastatic renal cell carcinoma, while producing both clinical and immunologic evidence of response.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendrites/immunology , Kidney Neoplasms/immunology , Aged , Cancer Vaccines , Carcinoma, Renal Cell/pathology , Female , Humans , Interleukin-2/therapeutic use , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging
18.
Int J Oncol ; 31(1): 193-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17549421

ABSTRACT

Adoptive T cell transfer after in vitro expansion represents an attractive cancer immunotherapy. The majority of studies so far have been focusing on the expansion of tumor infiltrated lymphocytes (TIL) and some have shown very encouraging results. Recently, we have developed a unique tumor immune response activator, dendritomas, by fusion of dendritic cells and tumor cells. Animal studies and early clinical trials have shown that dendritomas are able to activate tumor specific immune responses. In this study, we hypothesized that naïve T cells can be primed with dendritomas and expanded in vitro to develop an adoptive transfer therapy for patients who do not have solid tumors, such as leukemia. T cells were isolated and purified from lymph nodes of mice. The cells were then incubated with dendritomas made from syngeneic DCs and tumor cells and expanded in vitro using Dynabeads mouse CD3/CD28 T cell expander for approximately three weeks. The in vitro primed and expanded T cells showed tumor cell specific CTL activity and increased secretion of IFN-gamma. Tumor bearing mice receiving the in vitro expanded T cells survived significantly longer than control mice. Furthermore, the depletion of regulator T cells enhanced the survival of the mice that received the adoptive transfer therapy.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/transplantation , Animals , CD28 Antigens/analysis , CD3 Complex/analysis , Cell Fusion , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Mice, Inbred Strains , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Xenograft Model Antitumor Assays
19.
Int J Oncol ; 28(3): 585-93, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465362

ABSTRACT

A pilot clinical trial using dendritomas, purified hybrids from the fusion of dendritic/tumor cells combined with a low dose of IL-2, in metastatic melanoma patients was conducted in order to determine its safety and potential immunological and clinical responses. Ten metastatic melanoma patients were enrolled into this study. Dendritoma vaccines were created by fusing dendritic cells stained with green fluorescent dye with irradiated autologous tumor cells stained with red fluorescent dye and purifying the hybrids using immediate fluorescent-activated cell sorting. Initial vaccine was given subcutaneously and followed by IL-2 in serially elevated doses from 3-9 million units/m2 for 5 days. Repeated vaccinations were administered without IL-2, at 3-month intervals for a maximum of 5 times. Immune reactions were measured by the increase of interferon-gamma (IFN-gamma) expressing T cells. Vaccine doses ranged from 250,000 to 1,000,000 dendritomas. There was no grade 2 or higher toxicity directly attributable to the vaccine. All patients experienced toxicity due to IL-2 administration (9-grade 2, 3-grade 3, 1-grade 4). Eight of nine evaluable patients demonstrated immunologic reactions by increased IFN-gamma expressing T cells. One patient developed partial response at 12 weeks after the first vaccine. Nine months later, this patient achieved a complete response. In addition, two patients had stable disease for 9 and 4 months, respectively; one patient had a mixed response. Our findings demonstrated that dendritoma vaccines with a low dose of IL-2 can be safely administered to patients with metastatic melanoma and induce immunological and clinical responses.


Subject(s)
Dendritic Cells/immunology , Interleukin-2/therapeutic use , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/therapeutic use , Dendritic Cells/cytology , Female , Flow Cytometry , Humans , Hybrid Cells/immunology , Hybrid Cells/transplantation , Immunotherapy, Adoptive/methods , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Pilot Projects , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
20.
Biochem Biophys Res Commun ; 336(2): 430-7, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16137651

ABSTRACT

Insulin mimetics from natural sources are potential therapeutics that can act alone or supplement insulin and other anti-diabetic drugs in the prevention and treatment of diabetes. We recently reported the insulin-like glucose transport stimulatory activity of tannic acid (TA) in 3T3-L1 adipocytes. In this study, we find that chemically synthesized 1,2,3,4,6-penta-O-galloyl-beta-D-glucopyranose (beta-PGG), one of the components of TA, as well as its natural anomer alpha-PGG possess activity. Mechanistic studies in adipocytes with alpha-PGG, the more potent of the two anomers, reveal that inhibitors that block the insulin-mediated glucose transport, including one that inhibits the insulin receptor (IR), also completely abolish the glucose transport activated by alpha-PGG. In addition, alpha-PGG induces phosphorylation of the IR and Akt, activates PI 3-kinase, and stimulates membrane translocation of GLUT 4. Receptor binding studies indicate that alpha-PGG binds to the IR and affects the binding between insulin and IR by reducing the maximum binding of insulin to IR without significantly altering the binding affinity of insulin to IR. Western blotting analysis of the products of a cross-linking reaction suggests that alpha-PGG may bind to IR at a site located on the alpha-subunit of the receptor. Animal studies demonstrate that PGG reduces blood glucose levels and improves glucose tolerance in diabetic and obese animals. Our results suggest that PGG may serve as a model for the development of new types of anti-diabetic and anti-metabolic syndrome therapeutics.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Hydrolyzable Tannins/administration & dosage , Hydrolyzable Tannins/pharmacokinetics , Insulin/pharmacology , Receptor, Insulin/metabolism , Signal Transduction/physiology , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Dose-Response Relationship, Drug , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Mice , Protein Binding , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL