Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Immunother ; 38(5): 197-210, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25962108

ABSTRACT

Natural killer (NK) cells are promising effector cells for adjuvant immunotherapy of cancer. So far, several preclinical studies have shown the feasibility of gene-engineered NK cells, which upon expression of chimeric antigen receptors (CARs) are redirected to otherwise NK cell-resistant tumors. Yet, we reasoned that the efficiency of an immunotherapy using CAR-modified NK cells critically relies on efficient migration to the tumor site and might be improved by the engraftment of a receptor specific for a chemokine released by the tumor. On the basis of the DNAX-activation protein 12 (DAP12), a signaling adapter molecule involved in signal transduction of activating NK cell receptors, we constructed an epidermal growth factor variant III (EGFRvIII)-CAR, designated MR1.1-DAP12 which confers specific cytotoxicity of NK cell towards EGFRvIII glioblastoma cells in vitro and to established subcutaneous U87-MG tumor xenografts. So far, infusion of NK cells with expression of MR1.1-DAP12 caused a moderate but significantly delayed tumor growth and increased median survival time when compared with NK cells transduced with an ITAM-defective CAR. Notably, the further genetic engineering of these EGFRvIII-specific NK cells with the chemokine receptor CXCR4 conferred a specific chemotaxis to CXCL12/SDF-1α secreting U87-MG glioblastoma cells. Moreover, the administration of such NK cells resulted in complete tumor remission in a number of mice and a significantly increased survival when compared with the treatment of xenografts with NK cells expressing only the EGFRvIII-specific CAR or mock control. We conclude that chemokine receptor-engineered NK cells with concomitant expression of a tumor-specific CAR are a promising tool to improve adoptive tumor immunotherapy.


Subject(s)
ErbB Receptors/immunology , Gene Expression , Glioblastoma/etiology , Immunotherapy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, CXCR4/genetics , Animals , Cell Line , Cell- and Tissue-Based Therapy , Chemokine CXCL12/biosynthesis , Chemotaxis/genetics , Chemotaxis/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , Ectopic Gene Expression , Female , Gene Order , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Lentivirus/genetics , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, CXCR4/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic , Tumor Burden , Xenograft Model Antitumor Assays
2.
J Immunol ; 194(7): 3201-12, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25740942

ABSTRACT

NK cells are emerging as new effectors for immunotherapy of cancer. In particular, the genetic engraftment of chimeric Ag receptors (CARs) in NK cells is a promising strategy to redirect NK cells to otherwise NK cell-resistant tumor cells. On the basis of DNAX-activation protein 12 (DAP12), a signaling adaptor molecule involved in signal transduction of activating NK cell receptors, we generated a new type of CAR targeting the prostate stem cell Ag (PSCA). We demonstrate in this article that this CAR, designated anti-PSCA-DAP12, consisting of DAP12 fused to the anti-PSCA single-chain Ab fragment scFv(AM1) confers improved cytotoxicity to the NK cell line YTS against PSCA-positive tumor cells when compared with a CAR containing the CD3ζ signaling chain. Further analyses revealed phosphorylation of the DAP12-associated ZAP-70 kinase and IFN-γ release of CAR-engineered cells after contact with PSCA-positive target cells. YTS cells modified with DAP12 alone or with a CAR bearing a phosphorylation-defective ITAM were not activated. Notably, infused YTS cells armed with anti-PSCA-DAP12 caused delayed tumor xenograft growth and resulted in complete tumor eradication in a significant fraction of treated mice. The feasibility of the DAP12-based CAR was further tested in human primary NK cells and confers specific cytotoxicity against KIR/HLA-matched PSCA-positive tumor cells, which was further enhanced by KIR-HLA mismatches. We conclude that NK cells engineered with DAP12-based CARs are a promising tool for adoptive tumor immunotherapy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Proteins/genetics , Neoplasms/genetics , Neoplasms/immunology , Receptors, Natural Killer Cell/genetics , Recombinant Fusion Proteins , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Genetic Vectors/genetics , Humans , Immunophenotyping , Immunotherapy , Immunotherapy, Adoptive , Interferon-gamma/biosynthesis , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Xenograft Model Antitumor Assays , ZAP-70 Protein-Tyrosine Kinase/metabolism
3.
Prostate ; 74(13): 1347-58, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25053504

ABSTRACT

BACKGROUND: There is still a need for new therapeutic options against prostate cancer. Conventional single-chain bispecific antibodies (bsAbs), that directly cross-link T cells and tumor cells, hold great potential for efficient tumor treatment. However, rapid development of novel bsAbs is hampered by laborious optimization to improve their efficacy and reduce potential side effects. To accelerate the development of a novel antibody tool for the redirection of T cells to different tumor-associated antigens, we recently introduced a modular targeting system. METHODS: We here describe a novel modular system for treatment of prostate cancer by retargeting of T cells to the prostate stem cell antigen (PSCA). Functionality of the novel PSCA-specific modular system was investigated in vitro by T cell activation and chromium release assays as well as in immunodeficient mice. RESULTS: Similar to a conventional bsAb CD3-PSCA, the novel PSCA-specific modular system induces activation of both CD4+ and CD8+ T cells leading to secretion of pro-inflammatory cytokines and highly efficient target-specific tumor cell lysis. The novel TM was ready-to-use from the time point of construction and functional at low E:T ratios and picomolar concentrations without further optimization. In addition, the PSCA-specific modular system delays outgrowth of s.c. tumors in mice comparable to bsAb CD3-PSCA. CONCLUSIONS: We have developed a novel PSCA-specific modular system which triggers an efficient T cell-mediated killing of PSCA+ tumor cells in vitro and in vivo. The new Ab-based targeting strategy can functionally replace conventional bsAbs and allows a flexible redirection of T cells to different tumor-associated antigens.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasm Proteins/immunology , Prostatic Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , GPI-Linked Proteins/immunology , Humans , Immunotherapy , Male , Mice , Prostate/immunology , Prostate/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology
4.
Mol Cancer ; 13: 107, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24886358

ABSTRACT

BACKGROUND: Survivin, a member of the inhibitor of apoptosis (IAP) gene family, has a dual role in mitosis and in apoptosis. It is abundantly expressed in every human tumor, compared with normal tissues. During mitosis Survivin assembles with the chromosomal passenger complex and regulates chromosomal segregation. Here, we aim to explore whether interference with the mitotic function of Survivin is linked to p53-mediated G1 cell cycle arrest and affects chromosomal stability. METHODS: In this study, we used HCT116, SBC-2, and U87-MG and generated corresponding isogenic p53-deficient cells. Retroviral vectors were used to stably knockdown Survivin. The resulting phenotype, in particular the mechanisms of cell cycle arrest and of initiation of aneuploidy, were investigated by Western Blot analysis, confocal laser scan microscopy, proliferation assays, spectral karyotyping and RNAi. RESULTS: In all cell lines Survivin-RNAi did not induce instant apoptosis but caused polyplodization irrespective of p53 status. Strikingly, polyploidization after knockdown of Survivin resulted in merotelic kinetochore spindle assemblies, γH2AX-foci, and DNA damage response (DDR), which was accompanied by a transient p53-mediated G1-arrest. That p53 wild type cells specifically arrest due to DNA damage was shown by simultaneous inhibition of ATM and DNA-PK, which abolished induction of p21waf/cip. Cytogenetic analysis revealed chromosomal aberrations indicative for DNA double strand break repair by the mechanism of non-homologous end joining (NHEJ), only in Survivin-depleted cells. CONCLUSION: Our findings suggest that Survivin plays an essential role in proper amphitelic kinetochore-spindle assembly and that constraining Survivin's mitotic function results in polyploidy and aneuploidy which cannot be controlled by p53. Therefore, Survivin critically safeguards chromosomal stability independently from p53.


Subject(s)
Aneuploidy , DNA End-Joining Repair , Inhibitor of Apoptosis Proteins/genetics , Mitosis , Polyploidy , Tumor Suppressor Protein p53/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromosome Segregation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Gene Expression Regulation , Genomic Instability , Histones/genetics , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Karyotyping , Kinetochores/metabolism , Kinetochores/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Survivin , Tumor Suppressor Protein p53/metabolism
5.
PLoS One ; 9(4): e93745, 2014.
Article in English | MEDLINE | ID: mdl-24699869

ABSTRACT

Genetically engineered T lymphocytes are a promising option for cancer therapy. Prior to adoptive transfer they have to be expanded in vitro to reach therapeutically sufficient numbers. So far, no universal method exists for selective in vitro expansion of engineered T lymphocytes. In order to overcome this problem and for proof of concept we incorporated a novel unique peptide sequence of ten amino acids as epitope (E-Tag) into the binding domains of two novel chimeric antigen receptors (ECARs) directed against either prostate stem cell antigen (PSCA) for the treatment of prostate cancer (PCa) or CD33 for the treatment of acute myeloide leukemia (AML). The epitope tag then was utilized for expanding ECAR engrafted T cells by triggering the modified T cells via a monoclonal antibody directed against the E-Tag (Emab). Moreover, the E-Tag served as an efficient selection epitope for immunomagnetic isolation of modified T cells to high purity. ECAR engrafted T cells were fully functional and mediated profound anti-tumor effects in the respective models of PCa or AML both in vitro and in vivo. The method can be integrated straightforward into clinical protocols to improve therapeutic efficiency of tumor treatment with CAR modified T lymphocytes.


Subject(s)
Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Epitopes , Humans , Receptors, Antigen/genetics
6.
J Immunol ; 189(6): 3249-59, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22875801

ABSTRACT

Prostate cancer is the most common noncutaneous malignancy in men. The prostate stem cell Ag (PSCA) is a promising target for immunotherapy of advanced disease. Based on a novel mAb directed to PSCA, we established and compared a series of murine and humanized anti-CD3-anti-PSCA single-chain bispecific Abs. Their capability to redirect T cells for killing of tumor cells was analyzed. During these studies, we identified a novel bispecific humanized Ab that efficiently retargets T cells to tumor cells in a strictly Ag-dependent manner and at femtomolar concentrations. T cell activation, cytokine release, and lysis of target cells depend on a cross-linkage of redirected T cells with tumor cells, whereas binding of the anti-CD3 domain alone does not lead to an activation or cytokine release. Interestingly, both CD8+ and CD4+ T cells are activated in parallel and can efficiently mediate the lysis of tumor cells. However, the onset of killing via CD4+ T cells is delayed. Furthermore, redirecting T cells via the novel humanized bispecific Abs results in a delay of tumor growth in xenografted nude mice.


Subject(s)
Antibodies, Bispecific/physiology , Antibodies, Monoclonal, Humanized/physiology , Antigens, Neoplasm/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/immunology , Stem Cells/immunology , Tumor Stem Cell Assay , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Death/immunology , Epitopes, T-Lymphocyte/immunology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/immunology , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Prostatic Neoplasms/pathology , Stem Cells/pathology , Tumor Stem Cell Assay/methods
7.
Mol Ther ; 20(3): 572-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22186792

ABSTRACT

Using a mouse model we show that self-complementary (sc) adeno-associated virus (AAV) vectors pseudotyped with capsids of serotypes 2, 7 or 8 induce more potent transgene product-specific CD8(+) T cell and antibody responses compared to corresponding single-stranded (ss)AAV vectors. These data suggest that the higher and more rapidly appearing amounts of transgene product achieved with scAAV vectors may increase detrimental immune responses in gene transfer recipients.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Genome, Viral , Transgenes/immunology , Animals , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Dependovirus/immunology , Female , Gene Transfer Techniques , Genetic Vectors/immunology , Immunophenotyping , Kinetics , Mice , Mice, Inbred BALB C , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
8.
J Immunol ; 188(3): 1551-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22184723

ABSTRACT

Bispecific Abs hold great potential for immunotherapy of malignant diseases. Because the first components of this new drug class are now entering clinical trials, all aspects of their mode of action should be well understood. Several studies proved that CD8(+) and CD4(+) effector T cells can be successfully redirected and activated against tumor cells by bispecific Abs both in vitro and in vivo. To our knowledge, this study provides the first evidence that bispecific Abs can also redirect and activate regulatory T cells against a surface Ag, independently of their TCR specificity. After cross-linking, via a bispecific Ab, redirected regulatory T cells upregulate the activation markers CD69 and CD25, as well as regulatory T cell-associated markers, like CTLA-4 and FOXP3. The activated regulatory T cells secrete the immunosuppressive cytokine IL-10, but, in contrast to CD8(+) and CD4(+) effector T cells, almost no inflammatory cytokines. In addition, the redirected regulatory T cells are able to suppress effector functions of activated autologous CD4(+) T cells both in vitro and in vivo. Therefore, the potential risk for activation of regulatory T cells should be taken into consideration when bispecific Abs are applied for the treatment of malignant diseases. In contrast, an Ag/tissue-specific redirection of regulatory T cells with bispecific Abs holds great potential for the treatment of autoimmune diseases and graft rejection.


Subject(s)
Antibodies, Bispecific/therapeutic use , Molecular Targeted Therapy/methods , T-Lymphocytes, Regulatory/drug effects , Antibodies, Bispecific/pharmacology , Antigens, Surface/drug effects , Cell Line , Humans , Interleukin-10 , Lymphocyte Activation
9.
J Biomed Biotechnol ; 2011: 918471, 2011.
Article in English | MEDLINE | ID: mdl-22190859

ABSTRACT

An intact immune system is essential to prevent the development and progression of neoplastic cells in a process termed immune surveillance. During this process the innate and the adaptive immune systems closely cooperate and especially T cells play an important role to detect and eliminate tumor cells. Due to the mechanism of central tolerance the frequency of T cells displaying appropriate arranged tumor-peptide-specific-T-cell receptors is very low and their activation by professional antigen-presenting cells, such as dendritic cells, is frequently hampered by insufficient costimulation resulting in peripheral tolerance. In addition, inhibitory immune circuits can impair an efficient antitumoral response of reactive T cells. It also has been demonstrated that large tumor burden can promote a state of immunosuppression that in turn can facilitate neoplastic progression. Moreover, tumor cells, which mostly are genetically instable, can gain rescue mechanisms which further impair immune surveillance by T cells. Herein, we summarize the data on how tumor cells evade T-cell immune surveillance with the focus on solid tumors and describe approaches to improve anticancer capacity of T cells.


Subject(s)
Immunologic Surveillance , Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Escape , Animals , Dendritic Cells/immunology , Humans , Immune Tolerance , Neoplasm Proteins/immunology , Neoplasms/pathology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/pathology
10.
Neuro Oncol ; 13(10): 1074-89, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21788344

ABSTRACT

Malignant glioma represents the most common primary adult brain tumor in Western industrialized countries. Despite aggressive treatment modalities, the median survival duration for patients with glioblastoma multiforme (GBM), the highest grade malignant glioma, has not improved significantly over past decades. One promising approach to deal with GBM is the inactivation of proteins essential for survival or progression of glioma cells by means of RNA interference (RNAi) techniques. A likely candidate for an RNAi therapy of gliomas is the inhibitor of apoptosis protein survivin. Survivin is involved in 2 main cellular processes-cell division and inhibition of apoptosis. We show here that stable RNAi of survivin induced polyploidy, apoptosis, and impaired proliferation of human U343-MG, U373-MG, H4, and U87-MG cells and of primary glioblastoma cells. Proteome profiler arrays using U373-MG cells identified a novel set of differentially expressed genes upon RNAi-mediated survivin knockdown. In particular, the death receptor TRAIL R2/DR5 was strongly upregulated in survivin-depleted glioma cells, inducing an enhanced cytotoxic response of allogeneic human NK cells. Moreover, an experimental in vivo therapy using polyethylenimine (PEI)/siRNA complexes for survivin knockdown efficiently blocked tumor growth of established subcutaneous U373-MG tumors and enhanced survival of NMRI(nu/nu) mice orthopically transplanted with U87-MG cells. We conclude that survivin is functionally relevant in gliomas and that PEI-mediated exogenous delivery of siRNA targeting survivin is a promising strategy for glioblastoma therapy.


Subject(s)
Brain Neoplasms/genetics , Genetic Therapy/methods , Glioma/genetics , Inhibitor of Apoptosis Proteins/genetics , RNA, Small Interfering/pharmacology , Animals , Apoptosis/physiology , Blotting, Western , Brain Neoplasms/pathology , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Glioma/pathology , Humans , In Situ Nick-End Labeling , Mice , Protein Array Analysis , RNA Interference , Real-Time Polymerase Chain Reaction , Survivin , Transduction, Genetic , Xenograft Model Antitumor Assays
11.
J Immunol ; 184(10): 5475-84, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20410485

ABSTRACT

Immune responses diminish with age resulting in an increased susceptibility of the elderly to infectious agents and an inability to mount protective immune responses to vaccines. Immunosenescence affects multiple aspects of the immune system, including CD8(+) T cells, which control viral infections and are assumed to prevent the development of cancers. In this study, we tested if CD8(+) T cell responses in aged mice could be enhanced through a vaccine that concomitantly expresses Ag and a molecule that blocks an immunoinhibitory pathway. Specifically, we tested a vaccine based on a replication-defective chimpanzee-derived adenovirus vector expressing the nucleoprotein (NP) of influenza A virus as a fusion protein with the HSV type 1 glycoprotein D, which through binding to the herpes virus entry mediator, blocks the immunoinhibitory herpes virus entry mediator B and T lymphocyte attenuator/CD160 pathways. Our results show that the vaccine expressing a fusion protein of NP and glycoprotein D induces significantly higher NP-specific CD8(+) T cell responses in young and aged mice compared with the vaccine expressing NP only.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A virus/immunology , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Signal Transduction/immunology , Up-Regulation/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Epitopes, T-Lymphocyte/genetics , Female , Genetic Vectors/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/physiology , Recombinant Fusion Proteins/immunology , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...