Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Blood ; 117(26): 7174-84, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21540460

ABSTRACT

Donor lymphocyte infusions have been effective in patients with chronic myeloid leukemia (CML) relapsing after allogeneic stem cell transplantation, but their use is associated with the risk of graft-versus-host disease. We investigated the effects of prophylactic infusion of in vitro-generated donor T cells reactive against peptides derived from CML-associated antigens. Fourteen CML patients received conditioning therapy followed by CD34(+)-selected peripheral blood stem cells from matched siblings (n = 7) or unrelated (n = 7) donors. Donor-derived mature dendritic cells generated in vitro from CD14(+) monocytes were loaded with human leukocyte Ag-restricted peptides derived from PR1, WT1, and/or B-cell receptor-ABL and used to repetitively stimulate donor CD8(+) T cells in the presence of IL-2 and IL-7. Stimulated T cells were infused 28, 56, and 112 days after transplantation. Thirteen patients are alive and 7 remain in molecular remission (median follow-up, 45 months). Interestingly, all 4 patients receiving CD8(+) T cells displaying marked cytotoxic activity in vitro and detectable peptide-reactive CD8(+) T cells during follow-up have not experienced graft-versus-host disease or relapse. Our study reveals that prophylactic infusion of allogeneic CD8(+) T cells reactive against peptides derived from CML-associated antigens is a safe and promising therapeutic strategy. This trial was registered at www.clinicaltrials.gov as #NCT00460629.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/transplantation , Graft vs Host Disease/prevention & control , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Lymphocyte Depletion , Peptide Fragments/immunology , Peripheral Blood Stem Cell Transplantation/methods , Adult , Aged , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Cohort Studies , Dendritic Cells/metabolism , Feasibility Studies , Female , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Male , Middle Aged , Myeloblastin/metabolism , Peripheral Blood Stem Cell Transplantation/adverse effects , Remission Induction , Survival Analysis , Transplantation, Homologous , WT1 Proteins/metabolism , Young Adult
2.
PLoS One ; 6(1): e16315, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21283706

ABSTRACT

BACKGROUND: Previously, we identified a major myeloid-derived proinflammatory subpopulation of human blood dendritic cells which we termed slanDCs (e.g. Schäkel et al. (2006) Immunity 24, 767-777). The slan epitope is an O-linked sugar modification (6-sulfo LacNAc, slan) of P-selectin glycoprotein ligand-1 (PSGL-1). As slanDCs can induce neoantigen-specific CD4+ T cells and tumor-reactive CD8+ cytotoxic T cells, they appear as promising targets for an in vivo delivery of antigens for vaccination. However, tools for delivery of antigens to slanDCs were not available until now. Moreover, it is unknown whether or not antigens delivered via the slan epitope can be taken up, properly processed and presented by slanDCs to T cells. METHODOLOGY/PRINCIPAL FINDINGS: Single chain fragment variables were prepared from presently available decavalent monoclonal anti-slan IgM antibodies but failed to bind to slanDCs. Therefore, a novel multivalent anti-slanDC scaffold was developed which consists of two components: (i) a single chain bispecific recombinant diabody (scBsDb) that is directed on the one hand to the slan epitope and on the other hand to a novel peptide epitope tag, and (ii) modular (antigen-containing) linker peptides that are flanked at both their termini with at least one peptide epitope tag. Delivery of a Tetanus Toxin-derived antigen to slanDCs via such a scBsDb/antigen scaffold allowed us to recall autologous Tetanus-specific memory T cells. CONCLUSIONS/SIGNIFICANCE: In summary our data show that (i) the slan epitope can be used for delivery of antigens to this class of human-specific DCs, and (ii) antigens bound to the slan epitope can be taken up by slanDCs, processed and presented to T cells. Consequently, our novel modular scaffold system may be useful for the development of human vaccines.


Subject(s)
Amino Sugars/immunology , Antigens/administration & dosage , Dendritic Cells/immunology , Drug Delivery Systems/methods , Immunotherapy/methods , Antigen Presentation , Humans , Membrane Glycoproteins , T-Lymphocytes/immunology , Vaccines/immunology
3.
Haematologica ; 94(8): 1151-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19546436

ABSTRACT

Mesenchymal stem cells emerged as a promising treatment modality for steroid-refractory graft-versus-host disease, which represents a major complication of allogeneic hematopoietic stem cell transplantation. Dendritic cells (DCs) display an extraordinary capacity to induce T-cell responses and play a crucial role in the pathogenesis of graft-versus-host disease. Here, we investigated the impact of mesenchymal stem cells on the proinflammatory capacity of 6-sulfo LacNAc (slan) dendritic cells, representing a major subpopulation of human blood dendritic cells. Mesenchymal stem cells markedly impair maturation of slanDCs and their ability to secrete proinflammatory cytokines, which was dependent on prostaglandin E(2). In contrast, the release of anti-inflammatory IL-10 was improved by mesenchymal stem cells. Furthermore, mesenchymal stem cells efficiently inhibit slanDC-induced proliferation of CD4(+) and CD8(+) T cells and polarization of naïve CD4(+) T lymphocytes into Th1 cells. These results indicate that mesenchymal stem cells significantly impair the high proinflammatory capacity of slanDCs and further substantiate their potential for the treatment of graft-versus-host disease.


Subject(s)
Cytokines/metabolism , Dendritic Cells/metabolism , Mesenchymal Stem Cells/immunology , Amino Sugars/immunology , Antigens, CD/immunology , B7-2 Antigen/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Flow Cytometry , HLA-DR Antigens/immunology , Humans , Immunoglobulins/immunology , Interferon-gamma/metabolism , Interleukin-4/metabolism , Membrane Glycoproteins/immunology , Mesenchymal Stem Cells/cytology , CD83 Antigen
4.
J Virol ; 82(11): 5631-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367519

ABSTRACT

Here, we investigated the containment of virus replication in simian immunodeficiency virus (SIV) infection by CD8(+) lymphocytes. Escape mutations in Mamu-A*01 epitopes appeared first in SIV Tat TL8 and then in SIV Gag p11C. The appearance of escape mutations in SIV Gag p11C was coincident with compensatory changes outside of the epitope. Eliminating CD8(+) lymphocytes from rhesus monkeys during primary infection resulted in more rapid disease progression that was associated with preservation of canonical epitopes. These results confirm the importance of cytotoxic T cells in controlling viremia and the constraint on epitope sequences that require compensatory changes to go to fixation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Macaca mulatta/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , Gene Products, gag/blood , Gene Products, tat/blood , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Macaca mulatta/virology , Mutation/genetics , Simian Acquired Immunodeficiency Syndrome/virology
5.
J Virol ; 82(11): 5618-30, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367534

ABSTRACT

Previously we have shown that CD8(+) T cells are critical for containment of simian immunodeficiency virus (SIV) viremia and that rapid and profound depletion of CD4(+) T cells occurs in the intestinal tract of acutely infected macaques. To determine the impact of SIV-specific CD8(+) T-cell responses on the magnitude of the CD4(+) T-cell depletion, we investigated the effect of CD8(+) lymphocyte depletion during primary SIV infection on CD4(+) T-cell subsets and function in peripheral blood, lymph nodes, and intestinal tissues. In peripheral blood, CD8(+) lymphocyte-depletion changed the dynamics of CD4(+) T-cell loss, resulting in a more pronounced loss 2 weeks after infection, followed by a temporal rebound approximately 2 months after infection, when absolute numbers of CD4(+) T cells were restored to baseline levels. These CD4(+) T cells showed a markedly skewed phenotype, however, as there were decreased levels of memory cells in CD8(+) lymphocyte-depleted macaques compared to controls. In intestinal tissues and lymph nodes, we observed a significantly higher loss of CCR5(+) CD45RA(-) CD4(+) T cells in CD8(+) lymphocyte-depleted macaques than in controls, suggesting that these SIV-targeted CD4(+) T cells were eliminated more efficiently in CD8(+) lymphocyte-depleted animals. Also, CD8(+) lymphocyte depletion significantly affected the ability to generate SIV Gag-specific CD4(+) T-cell responses and neutralizing antibodies. These results reemphasize that SIV-specific CD8(+) T-cell responses are absolutely critical to initiate at least partial control of SIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Leukocyte Common Antigens/immunology , Macaca mulatta/immunology , Receptors, CCR5/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , Cell Movement/immunology , Disease Progression , Gene Products, gag/immunology , Intestines/cytology , Intestines/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/virology , Macaca mulatta/genetics , Macaca mulatta/metabolism , Macaca mulatta/virology , Male , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Survival Rate , Virus Replication
6.
Eur Urol ; 53(4): 694-708, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18061335

ABSTRACT

OBJECTIVES: The absence of effective therapies for advanced prostate cancer has entailed an intensive search for novel treatments. This review presents an overview of specific immunotherapeutic strategies for prostate cancer. METHODS: Current literature was reviewed regarding the identification of tumor antigens and the design of T-cell- and antibody-based immunotherapy for prostate cancer. The PubMed database was searched using the key words antibodies, clinical trials, dendritic cells, immunotherapy, prostate cancer, and T cells. RESULTS: T cells and antibodies are powerful components of the specific antitumor immune response. CD8+ cytotoxic T lymphocytes (CTLs) efficiently destroy tumor cells. CD4+ T cells improve the antigen-presenting capacity of dendritic cells (DCs) and support the stimulation of tumor-reactive CTLs. Monoclonal antibodies exhibit their antitumor effects via antibody-dependent cellular cytotoxicity and complement activation. Consequently, much attention has been given to the identification of tumor antigens that represent attractive targets for specific immunotherapy. Several prostate cancer-related antigens were described and used in clinical trials. Such studies were based on the administration of peptides, proteins, or DNA. Furthermore, men with prostate cancer were vaccinated with peptide-, protein-, or RNA-loaded DCs, which display an extraordinary capacity to induce tumor-reactive T cells. Monoclonal antibodies directed against surface antigens were also used. Clinical trials revealed that immunotherapeutic strategies represent safe and feasible concepts for the induction of immunologic and clinical responses in men with prostate cancer. CONCLUSIONS: Specific immunotherapy represents a promising treatment modality for prostate cancer. Further improvement of the current approaches is required and may be achieved by combining T-cell- and antibody-based vaccination strategies with radio-, hormone-, chemo-, or antiangiogenic therapy.


Subject(s)
Immunotherapy/trends , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Dendritic Cells/immunology , Humans , Male , Prostate-Specific Antigen/immunology , T-Lymphocytes/immunology
7.
Blood ; 110(8): 3078-81, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17616642

ABSTRACT

Donor dendritic cells (DCs) play a pivotal role in the induction of immunity and tolerance after peripheral blood stem cell transplantation (PBSCT). Treatment of healthy donors with granulocyte-colony stimulating factor (G-CSF) increases the numbers of tolerogenic DCs and T cells among mobilized blood leukocytes in the graft. SlanDCs (6-sulfo LacNAc+ DCs), a major source of IL-12 and TNF-alpha in blood, have not been studied in this respect. Here, we demonstrate that slanDCs (14.9 x 10(6)/L to 64.0 x 10(6)/L) are efficiently mobilized by G-CSF and retain their capacity to produce IL-12 and TNF-alpha at high levels. Furthermore, G-CSF-mobilized slanDCs programmed the differentiation of Th1 cells and displayed a particularly strong capacity to stimulate the proliferation of naive allogeneic T cells. Thus, slanDCs transfused into recipients of allogeneic peripheral blood stem cell (PBSC) transplants are functionally fully capable and may be critical in supporting graft-versus-host disease as well as graft-versus-leukemia effects.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Inflammation/immunology , Lipopolysaccharides/metabolism , Amino Sugars , Dendritic Cells/metabolism , Flow Cytometry , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Interleukin-12/biosynthesis , Transplantation Immunology , Tumor Necrosis Factor-alpha/biosynthesis
8.
Cancer Lett ; 245(1-2): 331-6, 2007 Jan 08.
Article in English | MEDLINE | ID: mdl-16504379

ABSTRACT

The development of T cell-based immunotherapies of cancer depends on the identification of tumor-associated antigens capable of eliciting tumor-directed cytotoxic T cell responses. In malignant glioma the number of well-defined target antigens for cytotoxic T lymphocytes (CTLs) is still very limited. Recently, we demonstrated the abundant and specific overexpression of the transcription factor SOX11 in malignant glioma. Here, we describe the SOX11-derived peptide LLRRYNVAKV which is capable of inducing human leukocyte antigen-A*0201-restricted and tumor-reactive CTLs. This novel CTL epitope may serve as an attractive candidate for a T cell-based immunotherapy of glioma.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Glioma/immunology , High Mobility Group Proteins/immunology , Amino Acid Sequence , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/chemistry , Glioma/pathology , HLA-A Antigens/immunology , HLA-A2 Antigen , High Mobility Group Proteins/chemistry , Humans , K562 Cells , Molecular Weight , Oligopeptides/chemistry , Oligopeptides/immunology , SOXC Transcription Factors
9.
Cancer Lett ; 250(2): 177-93, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17084966

ABSTRACT

Clinical studies have shown that nuclear expression of the inhibitor of apoptosis protein Survivin in tumor cells predicted a favorable prognosis whereas cytosolic-localized protein caused a decreased overall survival. Therefore Survivin's subcellular localization may be important for its anti-apoptotic capacity. To address this question, we investigated localization and function of Survivin in normal human lung fibroblasts (NHLFs) and HeLa tumor cells. NHLFs of early passages expressed Survivin in the nucleus and were highly sensitive to C2 ceramide, which induces the mitochondrial apoptotic pathway. In contrast, NHLFs at higher passages relocated Survivin to the cytosol and became more resistant to C2 ceramide. Blocking nuclear export of Survivin by leptomycin B in HeLa cells increased susceptibility to C2 ceramide. In addition, transduction of HeLa cells with Survivin fused to a nuclear localization signal augmented basal expression levels of p53 and Bax and enhanced sensitivity for intrinsic apoptosis. Those findings suggest that a predominant nuclear localization of Survivin increases the sensitivity for pro-apoptotic stimuli, whereas nuclear export enables Survivin to fulfill its inhibitor of apoptosis function. A therapeutic intervention which holds Survivin in the nucleus of tumor cells might improve cancer therapy.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Tumor Suppressor Protein p53/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Fluorescent Antibody Technique , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Lung/cytology , Lung/metabolism , Subcellular Fractions/metabolism , Survivin
10.
J Immunol ; 177(3): 1698-707, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16849479

ABSTRACT

In previous studies, we detected a frame shift mutation in the gene encoding the autoantigen La of a patient with systemic lupus erythematosus. The mutant La mRNA contains a premature termination codon. mRNAs that prematurely terminate translation should be eliminated by RNA quality control mechanisms. As we find Abs specific for the mutant La form in approximately 30% of sera from anti-La-positive patients, we expected that mutant La mRNAs circumvent RNA control and the expression of mutant La protein could become harmful. Indeed, real-time PCR, immunostaining, and immunoblotting data of mice transgenic for the mutant La form show that mutant La mRNAs are not repressed in these animals and are translated to mutant La protein. In addition to the mutant La protein, we detected a minor portion of native human La in the mutant La-transgenic mice. Therefore, ribosomal frame shifting may allow the mutant La mRNA to escape from RNA control. Interestingly, expression of the mutant La mRNA results in a lupus-like disease in the experimental mice. Consequently, escape of mutant La mRNA from RNA control can have two effects: it 1) results in the expression of an immunogenic (neo)epitope, and 2) predisposes to autoimmunity.


Subject(s)
Autoantibodies/biosynthesis , Autoantigens/genetics , Epitopes/genetics , RNA Stability/immunology , RNA, Messenger/metabolism , Ribonucleoproteins/genetics , 3T3 Cells , Adult , Amino Acid Sequence , Animals , Autoantibodies/blood , Autoantigens/biosynthesis , Autoantigens/immunology , Codon, Nonsense , Epitopes/blood , Epitopes/immunology , Female , Humans , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Male , Mice , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Protein Biosynthesis , RNA Stability/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/immunology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , SS-B Antigen
11.
Immunity ; 24(6): 767-777, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16782032

ABSTRACT

Early and high-level production of IL-12 is crucial for effective immune responses against pathogens. Up until now, the cells providing this initial IL-12 have remained elusive. Here we show that a subset of human blood dendritic cells (DC) is the principal and primary source of IL-12p70 when blood leukocytes are stimulated with the TLR4-ligand LPS or with CD40-ligand. These so-called slanDC are characterized by the 6-sulfo LacNAc modification of PSGL-1, which is identified by the mAb M-DC8. The IL-12 response of slanDC requires a few hours of in vitro maturation, which is completely blocked in the presence of erythrocytes. This inhibition of maturation depends on the expression of CD47 on erythrocytes and of its ligand SIRPalpha on DC. While strictly controlled in the blood by erythrocytes, the high IL-12- and TNF-alpha-producing capacity of slanDC in tissues may be critical in fighting off pathogens; if uncontrolled, it may lead to adverse inflammatory reactions.


Subject(s)
Dendritic Cells/immunology , Erythrocytes/immunology , Interleukin-12/metabolism , Lipopolysaccharides/immunology , Amino Sugars , Antigens, Differentiation/analysis , Antigens, Differentiation/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , CD40 Ligand/pharmacology , CD47 Antigen/analysis , CD47 Antigen/metabolism , Coculture Techniques , Dendritic Cells/drug effects , Humans , Ligands , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/immunology , Psoriasis/immunology , Psoriasis/pathology , Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , Th1 Cells/immunology , Toll-Like Receptor 4/agonists
12.
Prostate ; 66(8): 811-21, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16482569

ABSTRACT

BACKGROUND: Immunotherapies might represent promising alternatives for the treatment of patients with hormone-refractory prostate cancer (HRPC). In a Phase I clinical trial, we evaluated a vaccination with dendritic cells (DCs) loaded with a cocktail consisting of HLA-A*0201-restricted peptides derived from five different prostate cancer-associated antigens [prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA), survivin, prostein, transient receptor potential p8 (trp-p8)]. METHODS: Eight HRPC patients received a total of four vaccinations every other week. Clinical and immunological responses were monitored by the determination of the serum PSA levels and by enzyme linked immunospot (ELISPOT) analyses, respectively. RESULTS: Apart from local skin reactions no side effects were noted. One patient displayed a partial response (PR; PSA decrease >50%) and three other patients showed stable PSA values or decelerated PSA increases. In ELISPOT analyses, three of four PSA responders also showed antigen-specific CD8+ T-cell activation against prostein, survivin, and PSMA. CONCLUSIONS: The described protocol represents a safe and feasible concept for the induction of clinical and immunological responses. The application of a peptide cocktail-derived from different antigens as a novel treatment modality is supposed to allow for the genetic and biologic heterogeneity of PCa.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , HLA-A Antigens/therapeutic use , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Aged , Antigens, Surface/analysis , Antigens, Surface/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Drug Resistance, Neoplasm , Glutamate Carboxypeptidase II/analysis , Glutamate Carboxypeptidase II/immunology , HLA-A Antigens/chemistry , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Immunoenzyme Techniques , Immunomagnetic Separation , Immunotherapy, Active , Inhibitor of Apoptosis Proteins , Male , Membrane Proteins/analysis , Membrane Proteins/immunology , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/immunology , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/immunology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Survivin , TRPM Cation Channels/analysis , TRPM Cation Channels/immunology
13.
Cancer Lett ; 236(2): 229-38, 2006 May 18.
Article in English | MEDLINE | ID: mdl-16046056

ABSTRACT

Activation of immune defense mechanisms against tumor antigens appears to be a promising therapeutic option for advanced prostate cancer (PCa). Specific immunotherapy critically depends on target antigens that are selectively expressed in the tumorous and optional in the normal prostate tissue in sufficient amounts. Although several prostate antigens have been described and some have already been used in clinical trials, a detailed comparative evaluation of their tissue-specificity and expression levels is still lacking. We determined the transcript levels of eight prostate targets (PSA, PAP, PSCA, PSGR, Prostein, PSMA, AIbZIP, trp-p8) in 16 different tissues by quantitative PCR and calculated a tissue-specificity index (TSI) for each molecule. Besides a preferential expression in prostate for all targets, striking differences in the expression levels and TSI were revealed which may be important for the selection of appropriate antigens for immunotherapy of PCa.


Subject(s)
Antigens, Surface/analysis , Glutamate Carboxypeptidase II/analysis , Membrane Glycoproteins/analysis , Neoplasm Proteins/analysis , Prostate-Specific Antigen/analysis , Prostate/metabolism , Receptors, Odorant/analysis , Acid Phosphatase , Antigens, Neoplasm , Antigens, Surface/genetics , Antigens, Surface/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cyclic AMP Response Element-Binding Protein , GPI-Linked Proteins , Gene Expression , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Humans , Immunotherapy/methods , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Prostate/immunology , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/metabolism , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
14.
Cancer Lett ; 236(1): 95-104, 2006 May 08.
Article in English | MEDLINE | ID: mdl-15979782

ABSTRACT

Recently, we identified the novel protein D-GPCR (Dresden G protein-coupled receptor) which is selectively overexpressed in human prostate cancer (PCa) and belongs to the subfamily of odorant-like orphan GPCRs. Quantification of D-GPCR transcripts in paired malignant and non-malignant prostate tissues of 106 patients with primary PCa by real-time PCR demonstrated a significant up-regulation of this gene in tumor samples. Furthermore, its expression increases with higher tumor stages and grades. The evaluation of D-GPCR expression as a potential molecular tumor marker was performed by receiver-operating characteristic curve (ROC) analysis resulting in an area under the curve (AUC) value of 0.6452. Hence, the evaluation of D-GPCR as possible additive diagnostic tool and putative therapy target appears promising.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Neoplasm Staging , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/metabolism , Sensitivity and Specificity , Up-Regulation
15.
J Virol ; 79(13): 8131-41, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15956558

ABSTRACT

Although live attenuated vaccines can provide potent protection against simian immunodeficiency virus (SIV) and simian-human immunodeficiency virus challenges, the specific immune responses that confer this protection have not been determined. To test whether cellular immune responses mediated by CD8+ lymphocytes contribute to this vaccine-induced protection, we depleted rhesus macaques vaccinated with the live attenuated virus SIVmac239Delta3 of CD8+ lymphocytes and then challenged them with SIVmac251 by the intravenous route. While vaccination did not prevent infection with the pathogenic challenge virus, the postchallenge levels of virus in the plasmas of vaccinated control animals were significantly lower than those for unvaccinated animals. The depletion of CD8+ lymphocytes at the time of challenge resulted in virus levels in the plasma that were intermediate between those of the vaccinated and unvaccinated controls, suggesting that CD8+ cell-mediated immune responses contributed to protection. Interestingly, at the time of challenge, animals expressing the Mamu-A*01 major histocompatibility complex class I allele showed significantly higher frequencies of SIV-specific CD8+ T-cell responses and lower neutralizing antibody titers than those in Mamu-A*01- animals. Consistent with these findings, the depletion of CD8+ lymphocytes abrogated vaccine-induced protection, as judged by the peak postchallenge viremia, to a greater extent in Mamu-A*01+ than in Mamu-A*01- animals. The partial control of postchallenge viremia after CD8+ lymphocyte depletion suggests that both humoral and cellular immune responses induced by live attenuated SIV vaccines can contribute to protection against a pathogenic challenge and that the relative contribution of each of these responses to protection may be genetically determined.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Products, env/immunology , Lymphocyte Depletion , Retroviridae Proteins, Oncogenic/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/pathogenicity , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/blood , Immunophenotyping , Macaca mulatta , Sequence Deletion , Simian Immunodeficiency Virus/physiology , Viral Vaccines , Virus Replication/drug effects , Virus Replication/physiology
16.
J Immunol ; 174(7): 4127-34, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15778372

ABSTRACT

Dendritic cells (DCs) are characterized by their unique capacity for primary T cell activation, providing the opportunity for DC-based cancer vaccination protocols. Novel findings reveal that besides their role as potent inducers of tumor-specific T cells, human DCs display additional antitumor effects. Most of these data were obtained with monocyte-derived DCs, whereas studies investigating native blood DCs are limited. In the present study, we analyze the tumoricidal capacity of M-DC8(+) DCs, which represent a major subpopulation of human blood DCs. We demonstrate that IFN-gamma-stimulated M-DC8(+) DCs lyse different tumor cell lines but not normal cells. In addition, we show that tumor cells markedly enhance the production of TNF-alpha by M-DC8(+) DCs via cell-to-cell contact and that this molecule essentially contributes to the killing activity of M-DC8(+) DCs. Furthermore, we illustrate the ability of M-DC8(+) DCs to promote proliferation, IFN-gamma production, and tumor-directed cytotoxicity of NK cells. The M-DC8(+) DC-mediated enhancement of the tumoricidal potential of NK cells is mainly dependent on cell-to-cell contact. These results reveal that, in addition to their crucial role in activating tumor-specific T cells, blood DCs exhibit direct tumor cell killing and enhance the tumoricidal activity of NK cells. These findings point to the pivotal role of DCs in triggering innate and adaptive immune responses against tumors.


Subject(s)
Cytotoxicity, Immunologic , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Neoplasms/pathology , Blood Cells , CD8 Antigens , Cell Communication , Cell Line, Tumor , Dendritic Cells/cytology , Humans , Immunity , Lymphocyte Activation/immunology , Neoplasms/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/physiology
17.
Prostate ; 64(4): 387-400, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15761874

ABSTRACT

BACKGROUND: The understanding of the molecular biology of the prostate and the process of prostate carcinogenesis is brought forward by the identification and characterization of new genes specifically expressed in prostate tissue. The encoded proteins may, in addition, provide novel diagnostic and therapeutic tools in prostate carcinoma (PCa). Here, we identify the novel gene Dresden-transmembrane protein of the prostate (D-TMPP) that is overexpressed in human prostate and prostate cancer. METHODS: Proceeding from a prostate-specific expressed sequence tag identified with an Affymetrix chip-based expression database, the full-length cDNA of the novel gene was isolated from prostate tissue. The potential protein-coding function of the open reading frame (ORF) was tested by in vitro transcription-coupled translation and recombinant expression in transfected prostate cancer cells. The expression pattern of D-TMPP in malignant and nonmalignant tissues and tumor cell lines was analyzed by hybridization of a radioactively labeled cDNA probe with a multiple tissue expression array and by a quantitative real-time PCR assay. RESULTS: The D-TMPP-mRNA encodes a putative seven-span transmembrane protein of 883 amino acids and is selectively overexpressed in prostate tissue. D-TMPP represents the first cloned and characterized transcript of a family of eukaryotic genes. D-TMPP transcripts were detected in all analyzed pairs (n = 25) of malignant and nonmalignant prostate tissues. In the androgen-dependent PCa cell line LNCaP, D-TMPP was upregulated by methyltrienolone. CONCLUSIONS: We describe the novel prostate-restricted molecule D-TMPP widely expressed in prostate cancer tissues.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Multigene Family/genetics , Prostate/physiology , Prostatic Neoplasms/genetics , Aged , Amino Acid Sequence , Androgens/metabolism , Anoctamins , Base Sequence , DNA, Complementary/isolation & purification , Exons/genetics , Genome, Human , Humans , Male , Membrane Proteins/metabolism , Middle Aged , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics
18.
Biochem Biophys Res Commun ; 327(3): 765-73, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15649412

ABSTRACT

Survivin is critically involved in mitosis and when overexpressed enhances the activity of the Aurora B kinase, a serine-threonine kinase belonging to the family of oncogenic Aurora/IpI1p-related kinases. Both proteins interact with Ras GTPase-activating protein suggesting an impact on the Ras pathway. This study aimed at defining the role of survivin in proliferation and potential transformation of cells. When survivin was overexpressed in normal human lung fibroblasts, the characteristic track lanes of fibroblasts were disturbed and the rate of cell proliferation was increased. An enhanced level of p21(ras) mRNA and protein expression and concomitant rise in levels of activated p21(ras) were observed. Despite increased proliferation cell survival remained dependent on serum and cells were not able to form colonies in soft agar assays. These data suggest that overexpression of survivin increases cell growth but, despite the increase in active p21(ras), is not sufficient to transform primary cells. Yet, in addition to its anti-apoptotic function it might contribute to the accelerated growth of tumour cells by increasing p21(ras) activity.


Subject(s)
Cell Division/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Fibroblasts/metabolism , Microtubule-Associated Proteins/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins , Proto-Oncogene Proteins p21(ras)/genetics , Survivin , Transduction, Genetic , Tumor Cells, Cultured , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
19.
Oncol Rep ; 13(1): 139-44, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15583815

ABSTRACT

Malignant glioma comprises the majority of primary human brain tumors with 16,800 new cases reported each year in the USA. Its prognosis remains dismal despite numerous attempts to improve conventional therapeutic modalities. Therefore, much effort is devoted to the exploration of alternative forms of treatment such as immunotherapy. The identification of potential target structures highly overexpressed in brain tumors is a crucial prerequisite for the activation of the immune defense against malignant glioma cells. By screening an expression database for genes highly expressed in glioblastoma multiforme (GBM), we identified the Pit-Oct-Unc (POU) cooperating transcription factor SOX11 that is known to be crucially involved in brain development. Analysis of the expression pattern of SOX11 in different normal adult and fetal tissues by multiple tissue dot blot and by a highly sensitive quantitative PCR assay confirmed the selective overexpression of SOX11 in fetal brain tissue. Examination of tissue specimens obtained from malignant gliomas and from normal brain by quantitative real-time PCR (Q-RT-PCR) revealed upregulation of SOX11 in almost all tumor samples (15/16) as compared to the pooled normal brain. Seventy-five percent of the tumor samples (12/16) showed a 5- to more than 600-fold overexpression. We conclude that, after downregulation of SOX11 in the adult brain, its expression is reactivated during tumorigenesis and that SOX11 therefore represents a promising novel molecular target for adjuvant therapy of malignant gliomas.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , High Mobility Group Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/therapy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/therapy , High Mobility Group Proteins/genetics , High Mobility Group Proteins/immunology , Humans , Immunotherapy , Oligonucleotide Array Sequence Analysis , POU Domain Factors , SOXC Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics
20.
J Clin Endocrinol Metab ; 90(3): 1768-74, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15585555

ABSTRACT

A distinctive feature of malignant adrenocortical neoplasms is decreased major histocompatibility complex (MHC) class II molecule expression. However, it is unknown whether there exists a restriction to certain MHC genotypes and whether this involves alterations of the Fas/Fas ligand system and thereby affects tissue homeostasis. Therefore, MHC class II phenotype and genotype and expression patterns of the Fas/Fas ligand system were investigated in 24 adrenocortical tumors (n(Adenomas) = 14, n(Carcinomas) = 10) and an adrenal cancer cell line (NCI-H295). No MHC class II antigen expression was detected in carcinomas. The DRB1*01 genotype was found in 54.5% of patients with carcinoma (P = 0.046). No prevalence of any genotype could be detected in patients with adenomas, which exhibited varying levels of antigen expression. Fas receptor expression was 75.0% in adenomas compared with 20.0% in carcinomas (P = 0.0196), whereas ligand expression was 37.7% in adenomas and reached almost 100% in the carcinomas investigated in this study (P = 0.0033). In summary, the DRB1*01 genotype may be correlated to a higher risk for malignancy. Additional studies on MHC class II genotype and phenotype and the altered Fas/Fas ligand system in adrenal neoplasms may help to identify mechanisms of immune escape and suggest new diagnostic approaches.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , HLA-DR Antigens/genetics , Membrane Glycoproteins/metabolism , fas Receptor/metabolism , Adolescent , Adrenal Cortex Neoplasms/epidemiology , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/epidemiology , Adrenocortical Carcinoma/metabolism , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Fas Ligand Protein , Female , Genetic Predisposition to Disease/epidemiology , Genotype , HLA-DRB1 Chains , Humans , Immunohistochemistry , Male , Middle Aged , Prevalence , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...