Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Neoplasia ; 21(7): 653-664, 2019 07.
Article in English | MEDLINE | ID: mdl-31132676

ABSTRACT

The ubiquitin-proteasome system is elementary for cellular protein degradation and gained rising attention as a new target for cancer therapy due to promising clinical trials with bortezomib, the first-in class proteasome inhibitor meanwhile approved for multiple myeloma and mantle cell lymphoma. Both bortezomib and next-generation proteasome inhibitors mediate their effects by targeting the 20S core particle of the 26S proteasome. The novel small molecule inhibitor b-AP15 affects upstream elements of the ubiquitin-proteasome cascade by suppressing the deubiquitinase activity of both proteasomal regulatory 19S subunits and showed promising anticancer activity in preclinical models. Nonetheless, effects of inhibitors on the ubiquitin-proteasome system are not exclusively restricted to malignant cells: alteration of natural killer cell-mediated immune responses had already been described for drugs targeting either 19S or 20S proteasomal subunits. Moreover, it has been shown that bortezomib impairs dendritic cell (DC) phenotype and function at different levels. In the present study, we comparatively analyzed effects of bortezomib and b-AP15 on monocyte-derived DCs. In line with previous results, bortezomib exposure impaired maturation, antigen uptake, migration, cytokine secretion and immunostimulation, whereas treatment with b-AP15 had no compromising effects on these DC features. Our findings warrant the further investigation of b-AP15 as an alternative to clinically approved proteasome inhibitors in the therapy of malignancies, especially in the context of combinatorial treatment with DC-based immunotherapies.


Subject(s)
Deubiquitinating Enzymes/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Ubiquitin/genetics , Apoptosis/drug effects , Bortezomib/pharmacology , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/pathology , Deubiquitinating Enzymes/genetics , Humans , Monocytes/metabolism , Neoplasms/genetics , Neoplasms/pathology , Piperidones/pharmacology , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology
2.
Cancer Immunol Immunother ; 67(6): 935-947, 2018 06.
Article in English | MEDLINE | ID: mdl-29556699

ABSTRACT

The first therapeutic proteasome inhibitor bortezomib has clinical efficacy in mantle cell lymphoma (MCL) which resulted in its incorporation in treatment algorithms for this disease. Impairment of proteasomal function by bortezomib is mediated via inhibition of the 20S core particle. However, proteasome function can also be modified by targeting upstream components of the ubiquitin-proteasome system. Recently, b-AP15 has been identified as a small molecule achieving proteasome inhibition by targeting the deubiquitinase (DUB) activity of the 19S regulatory subunit and was found to inhibit cancer cell growth in preclinical analyses. In the present study, both direct antitumor effects and the possibility to induce natural killer group 2 member D ligands (NKG2DL) to reinforce NK cell immunity with b-AP15 were investigated to provide a rational basis for clinical evaluation of this novel DUB inhibitor in MCL. Treatment with b-AP15 resulted in reduced viability as well as induction of apoptosis in a time- and dose-dependent manner, which could be attributed to caspase activation in MCL cells. In addition, treatment with b-AP15 differentially induced NKG2DL expression and subsequent NK cell lysis of MCL cells. These results indicate that the DUB inhibitor b-AP15 displays substantial antitumor activity in human MCL and suggest that b-AP15 might be a novel therapeutic option in the treatment of MCL that warrants clinical investigation.


Subject(s)
Lymphoma, Mantle-Cell/genetics , Piperidones/therapeutic use , Proteinase Inhibitory Proteins, Secretory/therapeutic use , Apoptosis , Cell Line, Tumor , Humans , Killer Cells, Natural/metabolism , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Piperidones/pharmacology , Proteinase Inhibitory Proteins, Secretory/pharmacology
3.
Cancer Immunol Immunother ; 67(5): 775-783, 2018 May.
Article in English | MEDLINE | ID: mdl-29468363

ABSTRACT

In chronic myeloid leukemia (CML), the translocation t(9;22) results in the fusion protein BCR-ABL (breakpoint cluster region-abelson murine leukemia), a tyrosine kinase mediating oncogenic signaling which is successfully targeted by treatment with BCR-ABL inhibitors like imatinib. However, BCR-ABL inhibitors may also affect antitumor immunity. For instance, it was reported that imatinib impairs the function of dendritic cells (DCs) that play a central role in initiating and sustaining T cell responses. Meanwhile, second generation BCR-ABL inhibitors like nilotinib, which inhibits BCR-ABL with enhanced potency have become standard of treatment, at least in patients with BCR-ABL kinase domain mutations. In this study we analyzed the influence of therapeutic concentrations of nilotinib on human monocyte-derived DCs and compared its effects to imatinib. We found that both tyrosine kinase inhibitors (TKI) comparably and significantly impaired differentiation of monocytes to DCs as revealed by curtated downregulation of CD14 and reduced upregulation of CD1a and CD83. This was only partially restored after withdrawal of the TKI. Moreover, both TKI significantly reduced activation-induced IL-12p70 and C-C motif chemokine ligand (CCL) 3 secretion, while divergent TKI effects for CCL2 and CCL5 were observed. In contrast, only nilotinib significantly impaired the migratory capacity of DCs and their capacity to induce T-cell immune responses in MLRs. Our results indicate that imatinib and nilotinib may differ significantly with regard to their influence on antitumor immunity. Thus, for future combinatory approaches and particularly stop studies in CML treatment, choice of the most suitable BCR-ABL inhibitor requires careful consideration.


Subject(s)
Dendritic Cells/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imatinib Mesylate/pharmacology , Monocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Monocytes/cytology , Monocytes/immunology , Phenotype
5.
Clin Transl Immunology ; 4(11): e50, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26682057

ABSTRACT

Dendritic cells (DCs) arise from hematopoietic stem cells and develop into a discrete cellular lineage distinct from other leucocytes. Mainly three phenotypically and functionally distinct DC subsets are described in the human peripheral blood (PB): plasmacytoid DCs (pDCs), which express the key marker CD303 (BDCA-2), and two myeloid DC subsets (CD1c(+) DC (mDC1) and CD141(+) DC (mDC2)), which express the key markers CD1c (BDCA-1) and CD141 (BDCA-3), respectively. In addition to these primary cell subsets, DCs can also be generated in vitro from either CD34(+) stem/progenitor cells in the presence of Flt3 (Fms-related tyrosine kinase 3) ligand or from CD14(+) monocytes (monocyte-derived DCs (mo-DCs)) in the presence of granulocyte-macrophage colony-stimulating factor+interleukin-4 (GM-CSF+IL-4). Here we compare the reactivity patterns of HLDA10 antibodies (monoclonal antibody (mAb)) with pDCs, CD1c(+) DCs and CD141(+) DCs, as well as with CD14(+)-derived mo-DCs cultured for 7 days in the presence of 100 ng/ml GM-CSF plus 20 ng/ml IL-4. A detailed profiling of these DC subsets based on immunophenotyping and multicolour flow cytometry analysis is presented. Using the panel of HLDA10 Workshop mAb, we could verify known targets selectively expressed on discrete DC subsets including CD370 as a selective marker for CD141(+) DCs and CD366 as a marker for both myeloid subsets. In addition, vimentin and other markers are heterogeneously expressed on all three subsets, suggesting the existence of so far not identified DC subsets.

6.
Cell Commun Signal ; 13: 19, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25889792

ABSTRACT

BACKGROUND: Dendritic cells (DC) are the most potent antigen-presenting cells (APC) with the unique ability to activate naïve T cells and to initiate and maintain primary immune responses. Immunosuppressive and anti-inflammatory stimuli on DC such as the cytokine IL-10 suppress the activity of the transcription factor NF-κB what results in downregulation of costimulatory molecules, MHC and cytokine production. Glycoprotein NMB (GPNMB) is a transmembrane protein, which acts as a coinhibitory molecule strongly inhibiting T cell responses if present on APC. Interestingly, its expression on human monocyte-derived dendritic cells (moDC) is dramatically upregulated upon treatment with IL-10 but also by the BCR-ABL tyrosine kinase inhibitors (TKI) imatinib, nilotinib or dasatinib used for the treatment of chronic myeloid leukemia (CML). However, the molecular mechanisms responsible for GPNMB overexpression are yet unknown. RESULTS: The immunosuppressive cytokine IL-10 and the BCR-ABL TKI imatinib or nilotinib, that were examined here, concordantly inhibit the PI3K/Akt signaling pathway, thereby activating the downstream serine/threonine protein kinase GSK3ß, and subsequently the microphthalmia-associated transcription factor (MITF) that is phosphorylated and translocated into the nucleus. Treatment of moDC with a small molecule inhibitor of MITF activity reduced the expression of GPNMB at the level of mRNA and protein, indicating that GPNMB expression is in fact facilitated by MITF activation. In line with these findings, PI3K/Akt inhibition was found to result in GPNMB overexpression accompanied by reduced stimulatory capacity of moDC in mixed lymphocyte reactions (MLR) with allogeneic T cells that could be restored by addition of the GPNMB T cell ligand syndecan-4 (SD-4). CONCLUSIONS: In summary, imatinib, nilotinib or IL-10 congruently inhibit the PI3K/Akt signaling pathway thereby activating MITF in moDC, resulting in a tolerogenic phenotype. These findings extend current knowledge on the molecular mechanisms balancing activating and inhibitory signals in human DC and may facilitate the targeted manipulation of T cell responses in the context of DC-based immunotherapeutic interventions.


Subject(s)
Dendritic Cells/metabolism , Gene Expression Regulation/physiology , Membrane Glycoproteins/biosynthesis , Microphthalmia-Associated Transcription Factor/metabolism , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cells, Cultured , Dendritic Cells/cytology , Gene Expression Regulation/drug effects , Humans , Imatinib Mesylate , Interleukin-10/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology
7.
J Immunol ; 189(3): 1360-71, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22730533

ABSTRACT

Ligands of the prototypical activating NK receptor NKG2D render cancer cells susceptible to NK cell-mediated cytolysis if expressed at sufficiently high levels. However, malignant cells employ mechanisms to evade NKG2D-mediated immunosurveillance, such as NKG2D ligand (NKG2DL) shedding resulting in reduced surface expression levels. In addition, systemic downregulation of NKG2D on NK cells of cancer patients has been observed in many studies and was attributed to soluble NKG2DL (sNKG2DL), although there also are conflicting data. Likewise, relevant expression of NKG2DL in leukemia has been reported by some, but not all studies. Hence, we comprehensively studied expression, release, and function of the NKG2D ligands MHC class I chain-related molecules A and B and UL16-binding proteins 1-3 in 205 leukemia patients. Leukemia cells of most patients (75%) expressed at least one NKG2DL at the surface, and all investigated patient sera contained elevated sNKG2DL levels. Besides correlating NKG2DL levels with clinical data and outcome, we demonstrate that sNKG2DL in patient sera reduce NKG2D expression on NK cells, resulting in impaired antileukemia reactivity, which also critically depends on number and levels of surface-expressed NKG2DL. Together, we provide comprehensive data on the relevance of NKG2D/NKG2DL expression, release, and function for NK reactivity in leukemia, which exemplifies the mechanisms underlying NKG2D-mediated tumor immunosurveillance and escape.


Subject(s)
Immunologic Memory/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia/immunology , Leukemia/metabolism , Adult , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Down-Regulation/immunology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/pathology , Leukemia/pathology , Monitoring, Immunologic/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
8.
Cancer Immunol Immunother ; 61(2): 193-202, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21874302

ABSTRACT

Multiple approaches presently aim to combine targeted therapies using tyrosine kinase inhibitors with immunotherapy. Ex vivo-generated dendritic cells are frequently used in such strategies due to their unique ability to initiate primary T-cell immune responses. Besides governing tumor cell growth, many kinases targeted by tyrosine kinase inhibitors are involved in the development and function of dendritic cells and thus tyrosine kinase inhibitor therapy may cause immunoinhibitory side effects. We here report that exposure of developing human monocyte-derived dendritic cells to the BCR-ABL inhibitors imatinib, dasatinib, and nilotinib results in profound upregulation of the transmembrane glycoprotein osteoactivin that has recently been characterized as a negative regulator of T-cell activation. Thus, in line with osteoactivin upregulation, exposure to tyrosine kinase inhibitors resulted in significantly reduced stimulatory capacity of dendritic cells in mixed lymphocyte reactions that could be restored by the addition of blocking anti-osteoactivin antibody. Our data demonstrate that tyrosine kinase inhibitor-mediated inhibition of dendritic cell function is, at least in great part, mediated by upregulation of the immune inhibitory molecule osteoactivin.


Subject(s)
Dendritic Cells/drug effects , Immunotherapy , Membrane Glycoproteins/metabolism , T-Lymphocytes/metabolism , Up-Regulation , Antibodies, Monoclonal/metabolism , Benzamides , Cells, Cultured , Dasatinib , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Molecular Targeted Therapy , Piperazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thiazoles/pharmacology , Up-Regulation/drug effects
9.
Int J Cancer ; 127(9): 2119-28, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20143399

ABSTRACT

In chronic myeloid leukemia (CML), BCR/ABL-mediated oncogenic signaling can be targeted with the BCR/ABL-inhibitors Imatinib, Nilotinib and Dasatinib. However, these agents may also affect anti-tumor immunity. Here, we analyzed the effects of the 3 BCR/ABL-inhibitors on natural killer (NK) cell reactivity. Exposure of CML cells (K562, Meg-01) to pharmacological concentrations of Imatinib, Nilotinib and Dasatinib diminished expression of ligands for the activating immunoreceptor NKG2D to a similar extent. This resulted in comparably reduced NK cell cytotoxicity and IFN-gamma production. When direct effects on NK cell responses to K562 and primary CML cells as well as activating cytokines were studied, Dasatinib was found to abrogate NK cytotoxicity and cytokine production. Nilotinib did not alter cytotoxicity but, at high levels, impaired NK cytokine production, while Imatinib had no direct influence on NK cell reactivity. Of note, Nilotinib, but not the other BCR/ABL-inhibitors increased cell death within the preferentially cytokine-secreting CD56(bright)CD16(-) NK cell subset, which may, at least in part, serve to explain the effect of Nilotinib on NK cytokine production. Analysis of NK cell signaling revealed that Dasatinib inhibited proximal signaling events leading to decreased phosphorylation of PI3K and ERK that are crucial for NK cell reactivity. Imatinib and Nilotinib, in contrast, showed no relevant effect on NK cell PI3K or ERK activity. In light of the potential role of NK cells in the immunesurveillance of residual leukemia and for future combinatory immunotherapeutic approaches, our data indicate that choice and dosing of the most suitable BCR/ABL-inhibitor for a given patient require careful consideration.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Killer Cells, Natural/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Aluminum Silicates/metabolism , Benzamides , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Dasatinib , Down-Regulation , Histocompatibility Antigens Class I/metabolism , Humans , Imatinib Mesylate , Interferon-gamma/metabolism , K562 Cells , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/embryology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
10.
Int J Med Microbiol ; 300(1): 19-24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19781988

ABSTRACT

Dendritic cells (DCs) are key players in activation of the adaptive immune system by their ability of antigen presentation to and priming of T cells. An increasing body of evidence suggests that DCs may also play an important role in induction of tolerance, predominantly by induction of regulatory T cells (T(reg)). More recently, data have been published on how Toll-like receptor (TLR) ligands and cytokines affect DC differentiation, and how DC subsets might be involved in immunoregulation and tolerance rather than in T cell activation. The most important features of tolerance-inducing DCs appear to be their maturation state and their cytokine secretion pattern. The following types of tolerance-inducing DCs have been reported: immature DCs (DCs(im)) or DCs in the steady state (DCs(st)), DCs(IL-10), semi-mature DCs(TNF-alpha), semi-mature DCs(IL-6). With this review article we would like to discuss the aforementioned types of tolerogenic DCs with a focus on semi-mature DCs(IL-6) and discuss their potential role in maintenance of (hepatic or intestinal) immune homeostasis and inflammatory diseases such as inflammatory bowel disease.


Subject(s)
Dendritic Cells/immunology , Homeostasis , Immunomodulation , Interleukin-6/immunology , Toll-Like Receptors/immunology , Humans , Models, Biological
11.
Blood ; 115(15): 3058-69, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20008791

ABSTRACT

Natural killer (NK) cells play an important role in the immunosurveillance of leukemia. Their reactivity is governed by a balance of activating and inhibitory receptors including various members of the tumor necrosis factor receptor (TNFR) family. Here we report that human NK cells acquire expression of the TNFR family member CD137 upon activation, and NK cells of acute myeloid leukemia (AML) patients display an activated phenotype with substantial CD137 expression. CD137 ligand (CD137L) was detectable on leukemic cells in 35% of 65 investigated AML patients, but not on healthy CD34(+) cells, and expression was associated with monocytic differentiation. Bidirectional signaling following CD137-CD137L interaction induced the release of the immunomodulatory cytokines interleukin-10 and TNF by AML cells and directly diminished granule mobilization, cytotoxicity, and interferon-gamma production of human NK cells, which was restored by blocking CD137. Cocultures of NK cells with CD137L transfectants confirmed that human CD137 inhibits NK-cell reactivity, while activating signals were transduced by its counterpart on NK cells in mice. Our data underline the necessity to study the function of seemingly analog immunoregulatory molecules in mice compared with men and demonstrate that CD137-CD137L interaction enables immune evasion of AML cells by impairing NK-cell tumor surveillance in humans.


Subject(s)
4-1BB Ligand/immunology , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , 4-1BB Ligand/genetics , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cytokines/metabolism , Female , Gene Expression Regulation, Leukemic , Histocompatibility Antigens Class I/immunology , Humans , Immune Tolerance/immunology , Killer Cells, Natural/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lymphocyte Activation/immunology , Male , Mice , Middle Aged , Protein Binding , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Up-Regulation/genetics
12.
Blood ; 113(17): 4049-51, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19218548

ABSTRACT

Members of the caudal (cdx) family of homeobox proteins are essential regulators of embryonic blood development in zebrafish. Previously, we reported that the murine homologues (Cdx1, Cdx2, and Cdx4) affect formation and differentiation of embryonic stem cell (ESC)-derived hematopoietic progenitor cells. Consistent with the notion that embryonic pathways can reactivate during adult oncogenesis, recent studies suggest involvement of CDX2 in human acute myeloid leukemia (AML). Here we study CDX2 in healthy and leukemic human lymphoid cells, and show that a majority of leukemic samples display various degrees of aberrant CDX2 expression. Analysis of a cohort of 37 childhood acute lymphoblastic leukemia (ALL) patients treated in our hospital reveals that high CDX2 expression levels at diagnosis correlate with persistence of minimal residual disease (MRD) during the course of treatment. Thus, CDX2 expression levels may serve as a marker for adverse prognosis in pediatric ALL.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor , Case-Control Studies , Child , Homeodomain Proteins/genetics , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription, Genetic/genetics
13.
Clin Cancer Res ; 14(17): 5503-11, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18765542

ABSTRACT

PURPOSE: A prerequisite for the development of vaccination strategies is the identification and characterization of relevant tumor-associated antigen. Using microarray and reverse transcription-PCR analysis, we found matrix metalloproteinase (MMP)-7 to be extensively up-regulated in renal cell carcinomas and expressed in a broad variety of malignant cells. MMP-7 can promote cancer invasion and angiogenesis by proteolytic cleavage of extracellular matrix and basement membrane proteins, thus making it a promising target in the context of immunotherapies. EXPERIMENTAL DESIGN: To analyze the possible use of MMP-7 as a tumor-associated antigen, specific CTLs were induced using monocyte-derived dendritic cells electroporated with MMP-7-mRNA. In addition, to better characterize the fine specificity of these CTLs, MMP-7 MHC class I ligands were isolated and characterized in renal cell carcinoma tissue, which overexpressed MMP-7, by mass spectrometry-based peptide sequencing. Using this approach, we identified a novel HLA-A3-binding antigenic MMP-7 peptide. CTLs generated from healthy donors by in vitro priming with dendritic cells, pulsed with the novel peptide, were used as effectors in (51)Cr-release assays. RESULTS: The induced CTLs elicited an antigen-specific and HLA-restricted cytolytic activity against tumor cells endogenously expressing the MMP-7 protein. Furthermore, we were able to induce MMP-7-specific CTLs using peripheral blood mononuclear cells from a patient with acute lymphoblastic leukemia capable of recognizing the autologous leukemic blasts while sparing nonmalignant cells. CONCLUSIONS: Our study describes the identification of a novel broadly expressed T-cell epitope derived from the MMP-7 protein that represents an interesting candidate to be applied in immunotherapies of human malignancies targeting both tumor cells and neovascularization.


Subject(s)
Antigens, Neoplasm/isolation & purification , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Matrix Metalloproteinase 7/isolation & purification , Cell Line, Tumor , Electroporation , Epitopes , Epitopes, T-Lymphocyte/immunology , HLA-A3 Antigen/immunology , Humans , Matrix Metalloproteinase 7/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation
14.
Cancer Immunol Immunother ; 57(10): 1483-91, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18301890

ABSTRACT

Advances in tumor immunology and Identification of tumor-associated antigens (TAAs) provide a basis for the development of novel immunotherapies to treat malignant diseases. In order to identify novel TAAs, we performed comparative microarray analysis of (heterogeneous) tissues and found regulator of G protein-signaling 1 (RGS1) extensively up-regulated in renal cell carcinoma (RCC) tissues. To examine the possible function of this molecule as a novel, broadly applicable TAA, synthetic full-length RGS1-mRNA was synthesized for the transfection of monocyte-derived dendritic cells (DCs). These modified antigen-presenting cells (APCs) were then used to induce RGS1-specific cytotoxic T cells (CTLs) in vitro. The CTLs generated from several healthy donors and a patient with chronic lymphocytic leukemia (CLL) elicited an antigen-specific and HLA-A2- and -A3-restricted cytolytic activity against tumor cells endogenously expressing the RGS1 protein including renal cell carcinomas (RCCs), melanoma, ovarian carcinoma and the primary autologous CLL-blasts. In conclusion, our study demonstrates that the in vitro induction of RGS1-specific CTLs by RNA-transfected DCs is feasible and highly effective. Since this molecule is (over-) expressed in a broad variety of malignancies it might represent an interesting novel TAA in the context of cancer vaccines designed to target RGS1 expressing tumor cells.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , RGS Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/genetics , Cell Line, Tumor , Electroporation , Humans , Lymphocyte Activation/immunology , Oligonucleotide Array Sequence Analysis , RGS Proteins/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
15.
Blood ; 111(8): 4264-72, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-17698636

ABSTRACT

Human Dectin-1 (hDectin-1) is a member of the C-type lectin-like receptor family that was shown to be the major receptor for fungal beta-glucans and to play an important role in the cellular responses mediated by these carbohydrates. In this study, we demonstrate that hDectin-1 is involved in the uptake and cross-presentation of cellular antigens. Furthermore, activation of monocyte-derived dendritic cells (MDCs) with toll-like receptor 3 (TLR3) ligand but not with TLR2 ligand or TLR7 ligand resulted in down-regulation of hDectin-1 expression and reduced phagocytosis of apoptotic tumor cells as well as presentation of pp65-derived T-cell epitopes upon engulfment of cytomegalovirus (CMV)-infected human foreskin fibroblasts.


Subject(s)
Antigen Presentation/immunology , Cross-Priming/immunology , Membrane Proteins/immunology , Nerve Tissue Proteins/immunology , Apoptosis , Cell Line , Chromium , Cytomegalovirus Infections/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Fibroblasts/cytology , Fibroblasts/virology , Gene Expression Regulation , HLA-A2 Antigen/immunology , Humans , Lectins, C-Type , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phagocytosis , Phenotype , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptors/agonists
16.
Crit Rev Oncol Hematol ; 65(3): 191-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18055210

ABSTRACT

Cancer immunotherapy aims at eliciting an immune response directed against tumor antigens to help fight off residual tumor cells and thereby improve survival and quality of life of cancer patients. Different immunotherapeutic approaches share the use of dendritic cells (DCs) to present tumor-associated antigens to T-lymphocytes. Ex vivo generated DCs can be loaded with antigens and re-infused to the patients, or they can be used for ex vivo expansion of antitumor lymphocytes. Alternatively, methods exist to target antigens to DCs in vivo without need for ex vivo cell manipulations. The clinical studies have shown that DC administration to patients is safe and induces antigen-specific immunity. However, it seldom elicits objective clinical responses in patients with advanced-stage malignancies. Novel insights into DC and lymphocyte regulation are expected to lead to more effective vaccines in the near future. Meanwhile, efforts are directed at identifying the most appropriate clinical targets for active specific immunotherapies. Data suggests that vaccinations may indeed be beneficial when given in the adjuvant setting rather than to treat metastatic cancers. These issues are discussed here together with an overview of the DC-based antitumor immunotherapy studies.


Subject(s)
Dendritic Cells/immunology , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Separation , Humans , Immunotherapy
17.
J Biol Chem ; 282(52): 37836-43, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-17977821

ABSTRACT

The delivery of protein fragments to major histocompatibility complex (MHC)-loading compartments of professional antigen-presenting cells is essential in the adaptive immune response against pathogens. Apart from the crucial role of the transporter associated with antigen processing (TAP) for peptide loading of MHC class I molecules in the endoplasmic reticulum, TAP-independent translocation pathways have been proposed but not identified so far. Based on its overlapping substrate specificity with TAP, we herein investigated the ABC transporter ABCB9, also named TAP-like (TAPL). Remarkably, TAPL expression is strongly induced during differentiation of monocytes to dendritic cells and to macrophages. TAPL does not, however, restore MHC class I surface expression in TAP-deficient cells, demonstrating that TAPL alone or in combination with single TAP subunits does not form a functional transport complex required for peptide loading of MHC I in the endoplasmic reticulum. In fact, by using quantitative immunofluorescence and subcellular fractionation, TAPL was detected in the lysosomal compartment co-localizing with the lysosome-associated membrane protein LAMP-2. By in vitro assays, we demonstrate a TAPL-specific translocation of peptides into isolated lysosomes, which strictly requires ATP hydrolysis. These results suggest a mechanism by which antigenic peptides have access to the lysosomal compartment in professional antigen-presenting cells.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Dendritic Cells/cytology , Lysosomes/chemistry , Peptides/chemistry , Antigen Presentation , Antigen-Presenting Cells , Antigens/chemistry , Biological Transport , Cell Line, Tumor , Cloning, Molecular , Dendritic Cells/metabolism , HeLa Cells , Humans , Lipopolysaccharide Receptors/biosynthesis , Lysosomes/metabolism , Models, Biological , Monocytes/metabolism
18.
Clin Cancer Res ; 13(13): 3933-41, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17606727

ABSTRACT

PURPOSE: Histone deacetylases (HDAC) modulate gene transcription and chromatin assembly by modifying histones at the posttranscriptional level. HDAC inhibitors have promising antitumor activity and are presently explored in clinical studies. Cumulating evidence in animal models of immune disorders also suggests immunosuppressive properties for these small molecules, although the underlying mechanisms remain at present poorly understood. Here, we have evaluated the effects of two HDAC inhibitors currently in clinical use, sodium valproate and MS-275, on human monocyte-derived DCs. EXPERIMENTAL DESIGN: DCs were generated from monocytes through incubation with granulocyte macrophage colony-stimulating factor and interleukin-4. DC maturation was induced by addition of polyinosinic-polycytidylic acid. DC phenotype, immunostimulatory capacity, cytokine secretion, and migratory capacity were determined by flow cytometry, mixed leukocyte reaction, ELISA, and Transwell migration assay, respectively. Nuclear translocation of RelB, IFN regulatory factor (IRF)-3, and IRF-8 were determined by immunoblotting. RESULTS: HDAC inhibition skews DC differentiation by preventing the acquisition of the DC hallmark CD1a and by affecting the expression of costimulation and adhesion molecules. In addition, macrophage inflammatory protein-3beta/chemokine, motif CC, ligand 19-induced migration, immunostimulatory capacity, and cytokine secretion by DCs are also profoundly impaired. The observed defects in DC function on exposure to HDAC inhibitors seem to reflect the obstruction of signaling through nuclear factor-kappaB, IRF-3, and IRF-8. CONCLUSIONS: HDAC inhibitors exhibit strong immunomodulatory properties in human DCs. Our results support the evaluation of HDAC inhibitors in inflammatory and autoimmune disorders.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/enzymology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Active Transport, Cell Nucleus , Benzamides/pharmacology , Cell Differentiation , Cell Nucleus/metabolism , Dendritic Cells/immunology , Humans , Immune System , Immunosuppressive Agents/pharmacology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factors/metabolism , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Pyridines/pharmacology , Transcription Factor RelB/metabolism , Valproic Acid/pharmacology
19.
Clin Cancer Res ; 13(11): 3347-55, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545542

ABSTRACT

PURPOSE: Identification of tumor-associated antigens and advances in tumor immunology resulted in the development of vaccination strategies to treat patients with malignant diseases. In a novel experimental approach that combined comparative mRNA expression analysis of defined cell types with the characterization of MHC ligands by mass spectrometry, we found that regulator of G protein signaling 5 (RGS5) is extensively up-regulated in a broad variety of malignant cells, and we identified two HLA-A2- and HLA-A3-binding peptides derived from the RGS5 protein. Interestingly, RGS5 was recently shown to be involved in tumor angiogenesis. EXPERIMENTAL DESIGN: We used monocyte-derived dendritic cells pulsed with these novel antigenic peptides or transfected with RGS5-mRNA for the in vitro induction of CTLs, generated from healthy donors, to analyze the presentation of RGS5-deduced epitopes by malignant cells. RESULTS: The generated CTL lines elicited an antigen-specific and HLA-restricted cytolytic activity against tumor cells endogenously expressing the RGS5 protein. Furthermore, we were able to induce RGS5-specific CTLs using peripheral blood mononuclear cells from a patient with acute myeloid leukemia capable of recognizing the autologous leukemic blasts while sparing nonmalignant cells. CONCLUSIONS: These results indicate that the RGS5 peptides represent interesting candidates for the development of cancer vaccines designed to target malignant cells and tumor vessels.


Subject(s)
Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/chemistry , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , RGS Proteins/biosynthesis , RGS Proteins/immunology , Cancer Vaccines , Cell Line, Tumor , Dendritic Cells/metabolism , Gene Expression Regulation, Neoplastic , HLA-A2 Antigen/chemistry , HLA-A3 Antigen/chemistry , Humans , Leukemia, Myeloid, Acute/metabolism , Leukocytes, Mononuclear/metabolism , Monocytes/metabolism , Peptides/chemistry , RNA, Neoplasm/metabolism
20.
Cancer Res ; 67(11): 5489-97, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545631

ABSTRACT

Chronic myelogenous leukemia (CML) is a myeloproliferative disorder caused by excessive granulopoiesis due to the formation of the constitutively active tyrosine kinase BCR-ABL. An effective drug against CML is imatinib mesylate, a tyrosine kinase inhibitor acting on Abl kinases, c-KIT, and platelet-derived growth factor receptor. Recently, a study revealed that patients treated with imatinib showed impaired CTL responses compared with patients treated with IFN-alpha, which might be due to a treatment-induced reduction in immunogenicity of CML cells or immunosuppressive effects. In our study, we found that inhibition of BCR-ABL leads to a down-regulation of immunogenic antigens on the CML cells in response to imatinib treatment, which results in the inhibition of CML-directed immune responses. By treating CML cells with imatinib, we could show that the resulting inhibition of BCR-ABL leads to a decreased expression of tumor antigens, including survivin, adipophilin, hTERT, WT-1, Bcl-x(L), and Bcl-2 in correlation to a decreased development of CML-specific CTLs. In contrast, this reduction in immunogenicity was not observed when a CML cell line resistant to the inhibitory effects of imatinib was used, but could be confirmed by transfection with specific small interfering RNA against BCR-ABL or imatinib treatment of primary CML cells.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Piperazines/pharmacology , Pyrimidines/pharmacology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Benzamides , Dendritic Cells/immunology , Down-Regulation , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Lymphocyte Activation/drug effects , RNA, Small Interfering/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...