Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Surg Endosc ; 35(7): 3339-3353, 2021 07.
Article in English | MEDLINE | ID: mdl-32648038

ABSTRACT

INTRODUCTION: Endoscopic full-thickness resection (eFTR) using the full-thickness resection device (FTRD®) is a novel minimally invasive procedure that allows the resection of various lesions in the gastrointestinal tract including the colorectum. Real-world data outside of published studies are limited. The aim of this study was a detailed analysis of the outcomes of colonoscopic eFTR in different hospitals from different care levels in correlation with the number of endoscopists performing eFTR. MATERIAL AND METHODS: In this case series, the data of all patients who underwent eFTR between November 2014 and June 2019 (performed by a total of 22 endoscopists) in 7 hospitals were analyzed retrospectively regarding rates of technical success, R0 resection, and procedure-related complications. RESULTS: Colonoscopic eFTR was performed in 229 patients (64.6% men; average age 69.3 ± 10.3 years) mainly on the basis of the following indication: 69.9% difficult adenomas, 21.0% gastrointestinal adenocarcinomas, and 7.9% subepithelial tumors. The average size of the lesions was 16.3 mm. Technical success rate of eFTR was achieved in 83.8% (binominal confidence interval 78.4-88.4%). Overall, histologically complete resection (R0) was achieved in 77.2% (CI 69.8-83.6%) while histologically proven full-wall excidate was confirmed in 90.0% (CI 85.1-93.7%). Of the resectates obtained (n = 210), 190 were resected en bloc (90.5%). We did not observe a clear improvement of technical success and R0 resection rate over time by the performing endoscopists. Altogether, procedure-related complications were observed in 17.5% (mostly moderate) including 2 cases of acute gangrenous appendicitis requiring operation. DISCUSSION: In this pooled analysis, eFTR represents a feasible, effective, and safe minimally invasive endoscopic technique.


Subject(s)
Adenoma , Colonoscopy , Aged , Female , Hospitals , Humans , Male , Retrospective Studies , Treatment Outcome
3.
Cancer Immunol Immunother ; 63(4): 321-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24384835

ABSTRACT

BACKGROUND: Multiple studies have shown that dendritic cell (DC)-based vaccines can induce antitumor immunity. Previously, we reported that gemcitabine enhances the efficacy of DC vaccination in a mouse model of pancreatic carcinoma. The present study aimed at investigating the influence of gemcitabine on vaccine-induced anti-tumoral immune responses in a syngeneic pancreatic cancer model. MATERIALS AND METHODS: Subcutaneous or orthotopic pancreatic tumors were induced in C57BL/6 mice using Panc02 cells expressing the model antigen OVA. Bone marrow-derived DC were loaded with soluble OVA protein (OVA-DC). Animals received gemcitabine twice weekly. OVA-specific CD8(+) T-cells and antibody titers were monitored by FACS analysis and ELISA, respectively. RESULTS: Gemcitabine enhanced clinical efficacy of the OVA-DC vaccine. Interestingly, gemcitabine significantly suppressed the vaccine-induced frequency of antigen-specific CD8(+) T-cells and antibody titers. DC migration to draining lymph nodes and antigen cross-presentation were unaffected. Despite reduced numbers of tumor-reactive T-cells in peripheral blood, in vivo cytotoxicity assays revealed that cytotoxic T-cell (CTL)-mediated killing was preserved. In vitro assays revealed sensitization of tumor cells to CTL-mediated lysis by gemcitabine. In addition, gemcitabine facilitated recruitment of CD8(+) T-cells into tumors in DC-vaccinated mice. T- and B-cell suppression by gemcitabine could be avoided by starting chemotherapy after two cycles of DC vaccination. CONCLUSIONS: Gemcitabine enhances therapeutic efficacy of DC vaccination despite its negative influence on vaccine-induced T-cell proliferation. Quantitative analysis of tumor-reactive T-cells in peripheral blood may thus not predict vaccination success in the setting of concomitant chemotherapy.


Subject(s)
Adenocarcinoma/immunology , Antimetabolites, Antineoplastic/toxicity , B-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Deoxycytidine/analogs & derivatives , Immunosuppression Therapy , Pancreatic Neoplasms/immunology , Adenocarcinoma/drug therapy , Animals , Antibody Specificity , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Pancreatic Neoplasms/drug therapy , Peptide Fragments/immunology , Tumor Escape/drug effects , Gemcitabine
4.
PLoS One ; 8(2): e56867, 2013.
Article in English | MEDLINE | ID: mdl-23468885

ABSTRACT

OBJECTIVE: The specific inhibition of phosphodiesterase (PDE)4 and dual inhibition of PDE3 and PDE4 has been shown to decrease inflammation by suppression of pro-inflammatory cytokine synthesis. We examined the effect of roflumilast, a selective PDE4 inhibitor marketed for severe COPD, and the investigational compound pumafentrine, a dual PDE3/PDE4 inhibitor, in the preventive dextran sodium sulfate (DSS)-induced colitis model. METHODS: The clinical score, colon length, histologic score and colon cytokine production from mice with DSS-induced colitis (3.5% DSS in drinking water for 11 days) receiving either roflumilast (1 or 5 mg/kg body weight/d p.o.) or pumafentrine (1.5 or 5 mg/kg/d p.o.) were determined and compared to vehicle treated control mice. In the pumafentrine-treated animals, splenocytes were analyzed for interferon-γ (IFNγ) production and CD69 expression. RESULTS: Roflumilast treatment resulted in dose-dependent improvements of clinical score (weight loss, stool consistency and bleeding), colon length, and local tumor necrosis factor-α (TNFα) production in the colonic tissue. These findings, however, were not associated with an improvement of the histologic score. Administration of pumafentrine at 5 mg/kg/d alleviated the clinical score, the colon length shortening, and local TNFα production. In vitro stimulated splenocytes after in vivo treatment with pumafentrine showed a significantly lower state of activation and production of IFNγ compared to no treatment in vivo. CONCLUSIONS: These series of experiments document the ameliorating effect of roflumilast and pumafentrine on the clinical score and TNF expression of experimental colitis in mice.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Colitis/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Naphthyridines/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Aminopyridines/administration & dosage , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Benzamides/administration & dosage , Colitis/chemically induced , Colitis/drug therapy , Colon/drug effects , Colon/metabolism , Colon/pathology , Cyclopropanes/administration & dosage , Cyclopropanes/pharmacology , Dextran Sulfate/adverse effects , Disease Models, Animal , Female , Interferon-gamma/biosynthesis , Intestinal Mucosa/metabolism , Lectins, C-Type/metabolism , Mice , Naphthyridines/administration & dosage , Phosphodiesterase 3 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/administration & dosage , Spleen/cytology , Spleen/metabolism
5.
J Immunol ; 187(1): 55-63, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21613613

ABSTRACT

Cancer vaccines aim to induce CTL responses against tumors. Challenges for vaccine design are targeting Ag to dendritic cells (DCs) in vivo, facilitating cross-presentation, and conditioning the microenvironment for Th1 type immune responses. In this study, we report that ISCOM vaccines, which consist of ISCOMATRIX adjuvant and protein Ag, meet these challenges. Subcutaneous injection of an ISCOM vaccine in mice led to a substantial influx and activation of innate and adaptive immune effector cells in vaccine site-draining lymph nodes (VDLNs) as well as IFN-γ production by NK and NKT cells. Moreover, an ISCOM vaccine containing the model Ag OVA (OVA/ISCOM vaccine) was efficiently taken up by CD8α(+) DCs in VDLNs and induced their maturation and IL-12 production. Adoptive transfer of transgenic OT-I T cells revealed highly efficient cross-presentation of the OVA/ISCOM vaccine in vivo, whereas cross-presentation of soluble OVA was poor even at a 100-fold higher concentration. Cross-presenting activity was restricted to CD8α(+) DCs in VDLNs, whereas Langerin(+) DCs and CD8α(-) DCs were dispensable. Remarkably, compared with other adjuvant systems, the OVA/ISCOM vaccine induced a high frequency of OVA-specific CTLs capable of tumor cell killing in different tumor models. Thus, ISCOM vaccines combine potent immune activation with Ag delivery to CD8α(+) DCs in vivo for efficient induction of CTL responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cholesterol/administration & dosage , Cross-Priming/immunology , Cytotoxicity Tests, Immunologic/methods , Dendritic Cells/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Phospholipids/administration & dosage , Saponins/administration & dosage , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cells, Cultured , Cholesterol/immunology , Dendritic Cells/metabolism , Drug Combinations , Female , Gene Knock-In Techniques , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Phospholipids/immunology , Quillaja/immunology , Saponins/immunology
6.
Cancer Immunol Immunother ; 60(8): 1097-107, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21547597

ABSTRACT

BACKGROUND AND AIMS: Dendritic cell (DC)-based vaccination can induce antitumor T cell responses in vivo. This clinical pilot study examined feasibility and outcome of DC-based tumor vaccination for patients with advanced pancreatic adenocarcinoma. METHODS: Tumor lysate of patients with pancreatic carcinoma was generated by repeated freeze-thaw cycles of surgically obtained tissue specimens. Patients were eligible for DC vaccination after recurrence of pancreatic carcinoma or in a primarily palliative situation. DC were generated from peripheral blood mononuclear cells (PBMC), loaded with autologous tumor lysate, stimulated with TNF-α and PGE(2) and injected intradermally. All patients received concomitant chemotherapy with gemcitabine. Disease response was the primary endpoint. Individual immunological responses to DC vaccination were analyzed by T cell-based immunoassays using pre- and post-vaccination samples of non-adherent PBMC. RESULTS: Twelve patients received DC vaccination and concomitant chemotherapy. One patient developed a partial remission, and two patients remained in stable disease. Median survival was 10.5 months. No severe side effects were observed. Tumor-reactive T cells could be detected prior to vaccination. DC vaccination increased the frequency of tumor-reactive cells in all patients tested; however, the degree of this increase varied. To quantify the presence of tumor-reactive T cells, stimulatory indices (SI) were calculated as the ratio of proliferation-inducing capacity of lysate-loaded versus -unloaded DC. The patient with longest overall survival of 56 months had a high SI of 6.49, indicating that the presence of a pre-vaccination antitumor T cell response might be associated with prolonged survival. Five patients survived 1 year or more. CONCLUSION: DC-based vaccination can stimulate an antitumoral T cell response in patients with advanced or recurrent pancreatic carcinoma receiving concomitant gemcitabine treatment.


Subject(s)
Antigens, Neoplasm/metabolism , Cancer Vaccines , Carcinoma/therapy , Dendritic Cells/metabolism , Pancreatic Neoplasms/therapy , Adult , Aged , Antigen Presentation , Antigens, Neoplasm/immunology , Carcinoma/immunology , Carcinoma/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Dinoprostone/immunology , Dinoprostone/metabolism , Female , Humans , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pilot Projects , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Cancer ; 128(4): 897-907, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20473889

ABSTRACT

Vaccines based on immune stimulatory complexes (ISCOM) induce T-cell responses against tumor antigen (Ag). However, immune responses are impaired in pancreatic cancer patients. We investigated the efficacy of an ISCOM vaccine in a murine pancreatic carcinoma model. Panc02 cells expressing OVA as a model Ag were induced subcutaneously or orthotopically in the pancreas of C57BL/6 mice. Treatment consisted of an OVA containing ISCOM vaccine, either used alone or in combination with the TLR9 agonist CpG. The ISCOM vaccine effectively induced Ag-specific CTL capable of killing tumor cells. However, in mice with established tumors CTL induction by the vaccine was inefficient and did not affect tumor growth. Lack of efficacy correlated with increased numbers of Treg. Depletion of Treg with anti-CD25 mAb restored CTL induction and prolonged survival. Adding low-dose CpG to the ISCOM vaccine reduced Treg numbers, enhanced CTL responses and induced regression of pancreatic tumors in a CD8(+) T cell-dependent manner. Mice cured from the primary tumor mounted a memory T-cell response against wild-type Panc02 tumors, indicative of epitope spreading. Combining ISCOM vaccines with TLR agonists is a promising strategy for breaking tumor immune evasion and deserves further evaluation for the treatment of pancreatic carcinoma.


Subject(s)
Cancer Vaccines/therapeutic use , Disease Models, Animal , Immunotherapy , Oligodeoxyribonucleotides/therapeutic use , Pancreatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 9/agonists , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/prevention & control , Animals , Antigen-Antibody Complex/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immune Evasion , Immunization , Lymphatic Metastasis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/prevention & control , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
8.
J Immunol Methods ; 337(2): 97-105, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18657542

ABSTRACT

Phenotypical maturation, IL-12p70 production and migration upon chemokine receptor CCR7 ligation are currently proposed as requirements for the use of human monocyte-derived dendritic cells (DC) in antitumoral vaccination. We have previously described a short-term protocol for DC generation from monocytes including stimulation with TNF-alpha, IL-1beta and PGE(2) (FastDC). These "conventional" FastDC are mature, migrate in response to CCR7 ligation and effectively stimulate autologeous T cells in vitro, but are deficient in IL-12p70 production. Here, conventional FastDC were compared to FastDC activated with different TLR ligands. High levels of IL-12p70 were induced by combined activation of FastDC with TLR4 and TLR7/8 ligands. IL-12 secretion could be maximized by additional T cell-derived stimulation. However, TLR-stimulated FastDC failed to migrate upon CCR7 ligation, independent of additional activation with CD40 ligand and IFN-gamma. The presence of PGE(2) during TLR ligation fully restored migratory capacity of FastDC, but left IL-12p70 production and activation of tumor antigen-specific cytotoxic T cells unaffected, challenging previous findings obtained with standard 7-day monocyte-derived DC. The FastDC model thus not only represents an effective tool for antitumoral vaccination, but may also provide novel insights into human DC biology.


Subject(s)
Dendritic Cells/physiology , Dinoprostone/pharmacology , Interleukin-12/biosynthesis , Monocytes/cytology , Toll-Like Receptors/agonists , Antigens, Neoplasm/immunology , Cell Movement , Cells, Cultured , Humans , Interferon-gamma/biosynthesis , MART-1 Antigen , Neoplasm Proteins/immunology , Receptors, CCR7/physiology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptors/physiology
9.
Dig Dis Sci ; 52(7): 1642-52, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17393315

ABSTRACT

Previously we demonstrated an ameliorating effect of the interleukin-1beta converting enzyme (ICE) inhibitor pralnacasan on dextran sulfate sodium (DSS)-induced colitis. This study investigates the effects of pralnacasan on cytokine expression in DSS-induced colitis. Colitis was induced by oral administration of DSS. Mice were treated intraperitoneally with the ICE inhibitor pralnacasan (50 mg/kg body weight twice daily). Body weight as well as the presence of occult blood or diarrhea was monitored daily. Subgroups were sacrificed at days 4, 8, and 11 after the beginning of DSS application. Cytokine profiles in colonic tissue were analyzed on the protein level by ELISA and on the mRNA level by real time RT-PCR. Administration of DSS led to an increase in IL-18, IL-12, TNF-alpha, and IFN-gamma protein as well as IP-10 and TNF-alpha mRNA. The increase in IL-18 and IFN-gamma was reduced by ICE inhibition. Pralnacasan prevented DSS-induced colitis in C57BL/6 mice. In C57BL/6 mice, the DSS-induced increase in IP-10 mRNA, but not TNF-alpha mRNA, was completely prevented by ICE inhibition. In conclusion, prevention of colitis in C57BL/6 mice was associated with a suppresion of IP-10 mRNA, but not TNF-alpha mRNA expression, indicating that IL-18-mediated cytokine production is a key element in the pathogenesis of DSS-induced colitis.


Subject(s)
Caspase 1/metabolism , Chemokines, CXC/metabolism , Colitis/metabolism , Interferon-gamma/metabolism , Interleukin-18/metabolism , Animals , Azepines/therapeutic use , Caspase Inhibitors , Chemokine CCL2/metabolism , Chemokine CXCL1 , Chemokine CXCL10 , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Dextran Sulfate/toxicity , Epithelium/pathology , Female , Isoquinolines/therapeutic use , Mice , Mice, Inbred C57BL , Pyridazines/therapeutic use , RNA, Messenger/metabolism , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Leukoc Biol ; 80(2): 278-86, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16769767

ABSTRACT

Dendritic cells (DC) generated in vitro have to be viable and phenotypically mature to be capable of inducing T cell-mediated immunity after in vivo administration. To facilitate optimization of DC-based vaccination protocols, we investigated whether the cytokine environment and the mode of activation affect maturation and survival of DC derived from monocytes by a short-term protocol. Monocytes cultured for 24 h with granulocyte macrophage-colony stimulating factor and interleukin-4 were stimulated with proinflammatory mediators for another 36 h to generate mature DC. Additional activation with CD40 ligand and interferon (IFN)-gamma increased viability of DC and promoted definitive maturation as defined by maintenance of a mature phenotype after withdrawal of cytokines. Addition of IFN-alpha to DC cultures prior to stimulation further enhanced definitive maturation: IFN-alpha-primed DC expressed high levels of costimulatory molecules and CC chemokine receptor 7 (CCR7) up to 5 days after cytokine withdrawal. Compared with unprimed DC, IFN-alpha-primed DC displayed equal capacity to migrate upon CCR7 ligation and to prime antigen-specific T helper cell as well as cytolytic T cell responses. In conclusion, we show that optimal maturation and survival of monocyte-derived DC require multiple activation signals. Furthermore, we identified a novel role for IFN-alpha in DC development: IFN-alpha priming of monocytes promotes definitive maturation of DC upon activation.


Subject(s)
Dendritic Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon-alpha/pharmacology , Interferon-alpha/physiology , Interleukin-4/pharmacology , Monocytes/physiology , CD40 Ligand/pharmacology , Cell Differentiation , Cell Movement , Cells, Cultured , Humans , Interferon-gamma/pharmacology , Interleukin-12/blood , Monocytes/drug effects , Receptors, CCR7 , Receptors, Chemokine/immunology , T-Lymphocytes/physiology , T-Lymphocytes, Cytotoxic/physiology
11.
J Immunol Methods ; 302(1-2): 145-55, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15992809

ABSTRACT

Previously, we have shown that dendritic cells (DCs) with full T-cell stimulatory capacity can be derived from human monocytes after 48 h of in vitro culture (FastDC). Compared to a standard 7-day protocol, this new strategy not only reduces the time span and the amount of recombinant cytokines required, but may also resemble DC development in vivo more closely. Using a melanoma antigen model, we show here that FastDC prime CTL responses against tumor antigens as effectively as standard monocyte-derived DCs (moDCs). FastDC and moDCs derived from monocytes of HLA-A2(+) donors were loaded with the melanoma-associated, HLA-A(*)0201-restricted peptide Melan-A and cocultured with autologous CD3(+) T cells. After two weekly restimulations with freshly prepared, peptide-loaded FastDC or moDCs, binding of CD8(+) T cells to fluorescently labeled MHC-I/Melan-A-peptide complexes and intracellular cytokine staining revealed that the two DC preparations had an equal capacity to prime Melan-A-specific, IFN-gamma producing CD8(+) T cells. CTLs derived from cocultures with FastDC lysed Melan-A-loaded T2 cells even more effectively than CTLs primed by moDCs. Comparative analysis also revealed that FastDC possess an equal capacity to migrate in response to the chemokine receptor CCR-7 ligand 6Ckine. Importantly, DCs can be generated with higher yield and purity using the FastDC-protocol. The reliability and efficacy of this new strategy for DC development from monocytes may facilitate clinical investigation of DC-based tumor immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Cell Differentiation/immunology , Cytotoxicity Tests, Immunologic/methods , Dendritic Cells/immunology , Monocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Coculture Techniques , Cytokines/metabolism , Humans , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/metabolism
12.
J Immunother ; 28(4): 332-42, 2005.
Article in English | MEDLINE | ID: mdl-16000951

ABSTRACT

Dendritic cells (DCs) can induce cytotoxic T-cell (CTL) responses against tumor antigens in vitro and in vivo, yet few cancer patients experience tumor regression after DC-based vaccination. Combination with other treatment modalities, such as radiation or pharmacologic anticancer agents, may reduce tumor cell resistance against immune responses. The authors tested whether treatment with gemcitabine or cyclooxygenase-2 (COX-2) inhibitors increases the sensitivity of pancreatic carcinoma cells to CTL-mediated killing. Monocyte-derived DCs of HLA-A2+ donors were loaded with lysate from the HLA-A2+ pancreatic carcinoma cell line Panc-1 and co-cultured with autologous CD3+ T cells. ELISPOT and cytotoxicity assays performed after two rounds of in vitro stimulation confirmed induction of a tumor-specific CTL response. Changes in the magnitude and the effector mechanism of the CTL response were analyzed after treatment of Panc-1 cells with gemcitabine and COX-2 inhibitors. Compared with gemcitabine, COX-2 inhibitors more effectively sensitized Panc-1 cells to CTL-mediated killing and showed less inhibition of T-cell activation by DCs in vitro. Using anti-CD95 blocking antibody, the authors showed that the increase in CTL-mediated tumor cell killing observed after treatment with COX-2 inhibitors is dependent on CD95/CD95 ligand interaction. Increased apoptosis of Panc-1 cells treated with COX-2 inhibitor was also observed after incubation with agonistic anti-CD95 antibody. Sensitization of cancer cells to CD95-dependent killing by CTLs represents a novel mechanism of action for COX-2 inhibitors and provides a rationale for their concomitant use with immunotherapeutic strategies such as DC-based vaccination.


Subject(s)
Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coculture Techniques , Cyclooxygenase 2 , Cytokines/metabolism , Cytokines/pharmacology , Cytotoxicity, Immunologic/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Enzyme Inhibitors/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Membrane Proteins , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prostaglandin-Endoperoxide Synthases , Th1 Cells/immunology , fas Receptor/immunology , Gemcitabine
13.
J Pharmacol Exp Ther ; 308(2): 583-90, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14610233

ABSTRACT

The proinflammatory cytokines interleukin (IL)-1beta and IL-18 are supposed to play a crucial role in the pathogenesis of human inflammatory bowel disease. To exert biological activity, the precursors of both IL-1beta and IL-18 need to be cleaved by the interleukin-1beta-converting enzyme (ICE). IL-18 induces the synthesis of IFN-gamma in T cells and NK cells. In the present study, we investigated the effect of the specific ICE inhibitor pralnacasan in dextran sulfate sodium-induced murine colitis. Colitis was induced in BALB/c mice by 3.5% dextran sulfate sodium dissolved in drinking water for 10 days. Pralnacasan was administered either intraperitoneally or orally every day. To assess in vivo efficacy, a clinical disease activity score was evaluated daily. Colon length, expression of IL-18 in colonic tissue, expression of interferon-gamma (IFN-gamma) in paraaortal lymphocytes, and systemic production of IFN-gamma in splenocytes were analyzed post mortem. Intraperitoneally administered pralnacasan significantly reduced the clinical score compared with the dextran sulfate sodium control group from day 6 to day 10. Oral administration of pralnacasan also significantly reduced the clinical score at days 8 and 9. Administration of pralnacasan i.p. reduced the expression of intracolonic IL-18 significantly. Furthermore, pralnacasan reduced the number of IFN-gamma-positive lymphocytes in paraaortal lymph nodes. IFN-gamma synthesis in stimulated splenocytes was significantly suppressed in all pralnacasan-treated groups. No side effects of pralnacasan were observed. In conclusion, pralnacasan is effective in the prevention of dextran sulfate sodium-induced colitis. This effect is probably mediated by suppression of the proinflammatory cytokines IL-18, IL-1beta, and IFN-gamma.


Subject(s)
Azepines/pharmacology , Caspase Inhibitors , Colitis/pathology , Dextran Sulfate/pharmacology , Isoquinolines/pharmacology , Pyridazines/pharmacology , T-Lymphocytes, Helper-Inducer/drug effects , Animals , Azepines/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Colon/drug effects , Colon/pathology , Drug Interactions , Female , Interferon-gamma/metabolism , Interleukin-1 , Interleukin-18/metabolism , Isoquinolines/therapeutic use , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Para-Aortic Bodies , Pyridazines/therapeutic use , Spleen/pathology
14.
Biol Proced Online ; 5: 197-203, 2003.
Article in English | MEDLINE | ID: mdl-14615816

ABSTRACT

We developed a new 2-day protocol for the generation of dendritic cells (DCs) from human monocytes in vitro. First, we demonstrated that 24 hours of culture with GM-CSF and IL-4 are sufficient to generate immature DCs capable of antigen uptake. We then compared two different strategies for DC maturation: proinflammatory mediators were either added together with GM-CSF and IL-4 from the beginning of cell culture or added after 24 hours of differentiation with GM-CSF and IL-4. After 48 hours of total culture period, expression of activation markers was more pronounced in cells generated by the 2-step differentiation and activation method. Our new protocol for 2-day DC differentiation reduces labor, cost and time and also reliably renders high numbers of mature and viable DCs.

15.
Immunology ; 110(1): 38-47, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941139

ABSTRACT

Dendritic cells (DC) can be derived from monocytes in vitro by culture with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4). It is unknown whether this regimen reflects DC differentiation from blood precursors under physiological conditions. Induction of DC development from monocytes by interferon-alpha (IFN-alpha) may occur in vivo during infection or inflammation and thus may represent a more physiological approach to DC differentiation in vitro. Here, we show that incubation of GM-CSF-cultured monocytes with IFN-alpha does not induce DC differentiation: cells maintain their original phenotype and cytokine secretion pattern. Even after stimulation with pro-inflammatory or T-cell-derived activation signals, IFN-alpha-treated monocytes do not develop DC characteristics. Addition of IL-4 during stimulation of IFN-alpha-treated monocytes results in the rapid development of DC-like cells expressing co-stimulatory molecules, CD83 and chemokine receptor CCR7, indicating that some degree of developmental plasticity is preserved. However, DC pre-activated with IFN-alpha are less effective in inducing allogeneic or antigen-specific autologous T-cell proliferation, produce less IL-12 and express lower levels of CCR7 compared to DC generated by culture with GM-CSF and IL-4. Incubating GM-CSF-cultured monocytes simultaneously with IFN-alpha and IL-4 does not affect phenotypic maturation of DC, but reduces IL-12 production upon pro-inflammatory activation. We conclude that: (1) IFN-alpha fails to induce DC differentiation and thus cannot replace IL-4 in generating DC from monocytes in vitro; and (2) the presence of IFN-alpha prior to or during differentiation of DC from monocyte precursors alters their response to maturation stimuli and may affect their capacity to stimulate T helper type 1 immune responses in vivo.


Subject(s)
Dendritic Cells/immunology , Interferon-alpha/immunology , Lymphocyte Activation/immunology , Monocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Cytokines/biosynthesis , Dextrans/pharmacokinetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-12/metabolism , Interleukin-4/immunology , Lymphocyte Culture Test, Mixed , Monocytes/cytology , Monocytes/metabolism , Receptors, CCR7 , Receptors, Chemokine/metabolism , T-Lymphocytes/immunology
16.
J Immunol ; 170(8): 4069-76, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682236

ABSTRACT

It is widely believed that generation of mature dendritic cells (DCs) with full T cell stimulatory capacity from human monocytes in vitro requires 5-7 days of differentiation with GM-CSF and IL-4, followed by 2-3 days of activation. Here, we report a new strategy for differentiation and maturation of monocyte-derived DCs within only 48 h of in vitro culture. Monocytes acquire immature DC characteristics by day 2 of culture with GM-CSF and IL-4; they down-regulate CD14, increase dextran uptake, and respond to the inflammatory chemokine macrophage inflammatory protein-1alpha. To accelerate DC development and maturation, monocytes were incubated for 24 h with GM-CSF and IL-4, followed by activation with proinflammatory mediators for another 24 h (FastDC). FastDC expressed mature DC surface markers as well as chemokine receptor 7 and secreted IL-12 (p70) upon CD40 ligation in the presence of IFN-gamma. The increase in intracellular calcium in response to 6Ckine showed that chemokine receptor 7 expression was functional. When FastDC were compared with mature monocyte-derived DCs generated by a standard 7-day protocol, they were equally potent in inducing Ag-specific T cell proliferation and IFN-gamma production as well as in priming autologous naive T cells using tetanus toxoid as a model Ag. These findings indicate that FastDC are as effective as monocyte-derived DCs in stimulating primary, Ag-specific, Th 1-type immune responses. Generation of FastDC not only reduces labor, cost, and time required for in vitro DC development, but may also represent a model more closely resembling DC differentiation from monocytes in vivo.


Subject(s)
Cell Culture Techniques/methods , Dendritic Cells/cytology , Monocytes/cytology , Stem Cells/cytology , CD40 Ligand/pharmacology , Cell Differentiation/immunology , Cells, Cultured , Chemokine CCL21 , Chemokine CXCL12 , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dextrans/immunology , Dextrans/metabolism , Dinoprostone/pharmacology , Drug Combinations , Endocytosis/immunology , Epitopes, T-Lymphocyte/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Inflammation Mediators/pharmacology , Interferon-gamma/biosynthesis , Interleukin-1/pharmacology , Interleukin-12/biosynthesis , Interleukin-12/metabolism , Interleukin-12/physiology , Interleukin-4/pharmacology , Interphase/immunology , Ligands , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/metabolism , Receptors, CCR7 , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , Second Messenger Systems/immunology , Solubility , Stem Cells/immunology , Stem Cells/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocyte Subsets/immunology , Tetanus Toxoid/immunology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
17.
J Pharmacol Exp Ther ; 305(2): 549-56, 2003 May.
Article in English | MEDLINE | ID: mdl-12606674

ABSTRACT

Mesopram, a specific inhibitor of type-4 phosphodiesterase, decreases the synthesis of tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). In the present study, we investigated the effect of mesopram in dextran sulfate sodium (DSS)-induced murine colitis. In the preventive model, colitis was induced by DSS simultaneously with the application of mesopram in BALB/c mice. In the therapeutic model, colitis was induced in BALB/c mice by DSS over 7 days. At day 8, DSS was discontinued, and treatment was started. Mesopram was applied intraperitoneally or orally. The clinical score was calculated daily during the course of each study. Post mortem, colon length, histologic score, and expression of TNF-alpha and IFN-gamma in colons were determined. In the preventive model, mesopram significantly reduced the maximal clinical score, decreased colon shortening, and the histologic score. A dose finding study, using the preventive model, showed that most clinical and post mortem benefit was achieved with 50 mg/kg mesopram compared with 2 and 10 mg/kg. In the therapeutic model, i.p. mesopram treatment led to a significant reduction of clinical score. Both, i.p. and p.o. mesopram significantly reversed DSS-induced colon shortening and reduced the ex vivo colonic production of IFN-gamma. We conclude that the specific type-4 phosphodiesterase inhibitor mesopram ameliorates murine colitis both in a preventive and a therapeutic setting.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Colitis/drug therapy , Oxazoles/therapeutic use , Phosphodiesterase Inhibitors/therapeutic use , Aminosalicylic Acids/therapeutic use , Animals , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cytokines/biosynthesis , Dextran Sulfate , Dose-Response Relationship, Drug , Female , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/biosynthesis
18.
Cancer Res ; 62(8): 2347-52, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11956095

ABSTRACT

Tumor vaccines using dendritic cells (DCs) have been shown to induce antitumor CTL responses. The choice of the tumor antigen preparation used for DC loading is still an unresolved issue. We compared DCs pulsed with cell lysates, whole apoptotic tumor cells or their supernatants of the HLA-A2(+) human pancreatic carcinoma cell line Panc-1 for their capacity to activate T cells. Monocyte-derived DCs from HLA-A2(+) donors were pulsed with tumor antigen, matured subsequently, and cocultured with autologeous peripheral blood mononuclear cells. After three weekly restimulations with DCs, T-cell activation was assessed by intracellular IFN-gamma staining and cytotoxicity assays. Compared with lysate, pulsing DCs with the supernatant of apoptotic tumor cells induced a higher frequency of activated CTLs and T-helper cells, as well as an enhanced MHC class I-restricted tumor cell lysis. No activation of natural killer (NK) or gammadelta T cells was detected. Pulsing DCs with whole apoptotic tumor cells induced an even more pronounced lytic effect. However, in this case, MHC class-I blocking was only partially effective, and unrelated cell lines were also killed. IFN-gamma staining revealed activation of CTLs and T-helper cells, as well as NK and gammadelta T cells. Trans-well cultures of NK cells, apoptotic tumor cells, and DCs showed that NK cell activation was dependent on direct cell-to-cell contact with tumor cells and the presence of interleukin-12 produced by DCs. These results indicate that the choice of antigen preparation is a critical determinant in the induction of antitumor immunity. Tumor vaccines consisting of DCs and apoptotic tumor cells may be able to activate CTLs, as well as effector cells of the innate immune system.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Pancreatic Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Communication/immunology , Cytokines/biosynthesis , Cytokines/immunology , Humans , Immunotherapy, Adoptive/methods , Interleukin-1/biosynthesis , Interleukin-1/immunology , Lymphocyte Activation , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism
19.
Eur J Clin Pharmacol ; 58(1): 41-4, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11956672

ABSTRACT

OBJECTIVE: To develop a non-high-performance liquid chromatography method for the determination of thiopurine- S-methyltransferase (TPMT) phenotype using thin-layer chromatography and quantitative scanning. METHODS: TPMT reaction was performed using a radiochemical assay. The reaction product [(14)C]-6-methylmercaptopurine was separated using thin-layer chromatography and quantified by means of radioactive scanning. Day-to-day variance was determined to validate results. RESULTS: Determination of TPMT phenotype using thin-layer chromatography and quantitative scanning is reliable (day-to-day variance 8.5+/-1.7%, mean+/-SEM). Mean TPMT activity in 314 randomly selected patient samples was 11.8+/-3.3 units/ml red blood cells (mean+/-SD, range 3.5-25 units/ml). CONCLUSION: We developed a new assay variant for the determination of TPMT phenotype that is easy to perform, reliable and reduces production of radioactive waste. This may lead to more frequent pretreatment determination of TPMT phenotype and increase drug safety and efficacy by individualising thiopurine doses.


Subject(s)
Mercaptopurine/analogs & derivatives , Methyltransferases/genetics , Carbon Radioisotopes , Chromatography, Thin Layer/methods , Humans , Indicators and Reagents , Mercaptopurine/blood , Methyltransferases/blood , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...