Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 8(1): 17453, 2018 11 28.
Article in English | MEDLINE | ID: mdl-30487534

ABSTRACT

Bispecific T cell engager (BiTE) antibody constructs are successfully used as cancer therapeutics. We hypothesized that this treatment strategy could also be applicable for therapy of human cytomegalovirus (HCMV) infection, since HCMV-encoded proteins are abundantly expressed on the surface of infected cells. Here we show that a BiTE antibody construct directed against HCMV glycoprotein B (gB) and CD3 efficiently triggers T cells to secrete IFN-γ and TNF upon co-culture with fibroblasts infected with HCMV strain AD169, Towne or Toledo. Titration of gB expression levels in non-infected cells confirmed that already low levels of gB are sufficient for efficient triggering of T cells in presence of the BiTE antibody construct. Comparison of redirecting T cells with the bispecific antibody versus a chimeric antigen receptor (CAR) based on the same scFv showed a similar sensitivity for gB expression. Although lysis of infected target cells was absent, the BiTE antibody construct inhibited HCMV replication by triggering cytokine production. Notably, even strongly diluted supernatants of the activated T cells efficiently blocked the replication of HCMV in infected primary fibroblasts. In summary, our data prove the functionality of the first BiTE antibody construct targeting an HCMV-encoded glycoprotein for inhibiting HCMV replication in infected cells.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Viral/immunology , CD3 Complex/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Viral/pharmacology , Antibody Specificity/immunology , CD3 Complex/antagonists & inhibitors , Cytokines/metabolism , Cytomegalovirus/drug effects , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Humans , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Virus Replication/immunology
2.
J Transl Med ; 16(1): 26, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422056

ABSTRACT

BACKGROUND: During infection with human cytomegalovirus (HCMV) several viral proteins occur on cell surfaces in high quantity. We thus pursue an HLA-independent approach for immunotherapy of HCMV using chimeric antigen receptors (CARs) and bispecific BiTE® antibody constructs. In this context, HCMV-encoded proteins that mediate viral immune evasion and bind human IgG might represent particularly attractive target antigens. Unlike in observations of similar approaches for HIV and hepatitis B and C viruses, however, HCMV-infected cells develop a striking resistance to cytotoxic effector functions at later stages of the replication cycle. In our study we therefore wanted to test two hypotheses: (1) CAR T cells can efficiently inhibit HCMV replication independently from cytotoxic effector functions, and (2) HCMV can be targeted by CH2-CH3 IgG spacer domains that contain mutations previously reported to prevent exhaustion and to rescue CAR T cell function in vivo. METHODS: Replication of GFP-encoding recombinant HCMV in fibroblasts in the presence and absence of supernatants from T cell co-cultures plus/minus cytokine neutralizing antibodies was analyzed by flow cytometry. CARs with wild type and mutated CH2-CH3 domains were expressed in human T cells by mRNA electroporation, and the function of the CARs was assessed by quantifying T cell cytokine secretion. RESULTS: We confirm and extend previous evidence of antiviral cytokine effects and demonstrate that CAR T cells strongly block HCMV replication in fibroblasts mainly by combined secretion of IFN-γ and TNF. Furthermore, we show that fibroblasts infected with HCMV strains AD169 and Towne starting from day 3 have a high capacity for binding of human IgG1 and also strongly activate T cells expressing a CAR with CH2-CH3 domain. Importantly, we further show that mutations in the CH2-CH3 domain of IgG1 and IgG4, which were previously reported to rescue CAR T cell function by abrogating interaction with endogenous Fc receptors (FcRs), still enable recognition of FcRs encoded by HCMV. CONCLUSIONS: Our findings identify HCMV-encoded FcRs as an attractive additional target for HCMV immunotherapy by CARs and possibly bispecific antibodies. The use of specifically mutated IgG domains that bind to HCMV-FcRs without recognizing endogenous FcRs may supersede screening for novel binders directed against individual HCMV-FcRs.


Subject(s)
Cytomegalovirus/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Mutation/genetics , Receptors, Chimeric Antigen/metabolism , Receptors, Fc/metabolism , Cell Death , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Interferon-gamma/metabolism , Male , Protein Domains , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Viral Envelope Proteins , Virus Replication
3.
Front Microbiol ; 7: 844, 2016.
Article in English | MEDLINE | ID: mdl-27375569

ABSTRACT

In order to explore the potential of HLA-independent T cell therapy for human cytomegalovirus (HCMV) infections, we developed a chimeric antigen receptor (CAR) directed against the HCMV encoded glycoprotein B (gB), which is expressed at high levels on the surface of infected cells. T cells engineered with this anti-gB CAR recognized HCMV-infected cells and released cytokines and cytotoxic granules. Unexpectedly, and in contrast to analogous approaches for HIV, Hepatitis B or Hepatitis C virus, we found that HCMV-infected cells were resistant to killing by the CAR-modified T cells. In order to elucidate whether this phenomenon was restricted to the use of CARs, we extended our experiments to T cell receptor (TCR)-mediated recognition of infected cells. To this end we infected fibroblasts with HCMV-strains deficient in viral inhibitors of antigenic peptide presentation and targeted these HLA-class I expressing peptide-loaded infected cells with peptide-specific cytotoxic T cells (CTLs). Despite strong degranulation and cytokine production by the T cells, we again found significant inhibition of lysis of HCMV-infected cells. Impairment of cell lysis became detectable 1 day after HCMV infection and gradually increased during the following 3 days. We thus postulate that viral anti-apoptotic factors, known to inhibit suicide of infected host cells, have evolved additional functions to directly abrogate T cell cytotoxicity. In line with this hypothesis, CAR-T cell cytotoxicity was strongly inhibited in non-infected fibroblasts by expression of the HCMV-protein UL37x1, and even more so by additional expression of UL36. Our data extend the current knowledge on Betaherpesviral evasion from T cell immunity and show for the first time that, beyond impaired antigen presentation, infected cells are efficiently protected by direct blockade of cytotoxic effector functions through viral proteins.

4.
PLoS One ; 7(9): e44358, 2012.
Article in English | MEDLINE | ID: mdl-22970203

ABSTRACT

The Rho GTPases Rac1 and Cdc42 regulate a variety of cellular functions by signaling to different signal pathways. It is believed that the presence of a specific effector at the location of GTPase activation determines the route of downstream signaling. We previously reported about EGF-induced Ser-71 phosphorylation of Rac1/Cdc42. By using the phosphomimetic S71E-mutants of Rac1 and Cdc42 we investigated the impact of Ser-71 phosphorylation on binding to selected effector proteins. Binding of the constitutively active (Q61L) variants of Rac1 and Cdc42 to their specific interaction partners Sra-1 and N-WASP, respectively, as well as to their common effector protein PAK was abrogated when Ser-71 was exchanged to glutamate as phosphomimetic substitution. Interaction with their common effector proteins IQGAP1/2/3 or MRCK alpha was, however, hardly affected. This ambivalent behaviour was obvious in functional assays. In contrast to Rac1 Q61L, phosphomimetic Rac1 Q61L/S71E was not able to induce increased membrane ruffling. Instead, Rac1 Q61L/S71E allowed filopodia formation, which is in accordance with abrogation of the dominant Sra-1/Wave signalling pathway. In addition, in contrast to Rac1 transfected cells Rac1 S71E failed to activate PAK1/2. On the other hand, Rac1 Q61L/S71E was as effective in activation of NF-kappaB as Rac1 Q61L, illustrating positive signal transduction of phosphorylated Rac1. Together, these data suggest that phosphorylation of Rac1 and Cdc42 at serine-71 represents a reversible mechanism to shift specificity of GTPase/effector coupling, and to preferentially address selected downstream pathways.


Subject(s)
Neuropeptides/metabolism , Phosphoserine/metabolism , Signal Transduction , rac GTP-Binding Proteins/metabolism , Animals , Enzyme Activation , HEK293 Cells , Humans , Mice , Mutant Proteins/metabolism , NF-kappa B/metabolism , Neuropeptides/deficiency , Phenotype , Phosphorylation , Protein Binding , Pseudopodia/metabolism , Structure-Activity Relationship , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/deficiency , rac1 GTP-Binding Protein
5.
J Immunol ; 188(10): 4810-8, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22504646

ABSTRACT

The life span of dendritic cells (DCs) is determined by the balance of pro- and antiapoptotic proteins. In this study, we report that serum-free cultured human monocyte-derived DCs after TLR stimulation with polyinosinic acid-polycytidylic acid or LPS underwent apoptosis, which was correlated with low TNF production. Apoptosis was prevented by the addition of exogenous TNF or by concomitant stimulation with R-848, which strongly amplified endogenous TNF production. Neutralization of TNF confirmed that DC survival was mediated by autocrine TNF induced either by stimulation with R-848 or by ligation of CD40. DCs stimulated by polyinosinic acid-polycytidylic acid or IFN-ß, another known inducer of DC apoptosis, were characterized by high levels and activation of the proapoptotic protein BAK. The ratio of antiapoptotic BCL-2 to BAK correlated best with the survival of activated DCs. Addition of TNF increased this ratio but had little effect on BAX and XIAP. Knockdown experiments using small interfering RNAs confirmed that the survival of activated and also of immature DCs was regulated by BAK and showed that TNF was protective only in the presence of FLIP(L). Together, our data demonstrate that the survival of DCs during differentiation and activation depends on autocrine TNF and that the inhibition of BAK plays an important role in this process.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Autocrine Communication/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/physiology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/biosynthesis , CASP8 and FADD-Like Apoptosis Regulating Protein/biosynthesis , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Cell Count , Cell Differentiation/immunology , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/metabolism , Humans , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/biosynthesis , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/biosynthesis
6.
PLoS One ; 7(2): e31210, 2012.
Article in English | MEDLINE | ID: mdl-22355347

ABSTRACT

We explored the possibility to target Ewing's sarcoma family of tumors (ESFT) by redirecting T cells. To this aim, we considered NKG2D-ligands (NKG2D-Ls) as possible target antigens. Detailed analysis of the expression of MICA, MICB, ULBP-1, -2, and -3 in fourteen ESFT cell lines revealed consistent expression of at least one NKG2D-L. Thus, for redirecting T cells, we fused a CD3ζ/CD28-derived signaling domain to the ectodomain of NKG2D, however, opposite transmembrane orientation of this signaling domain and NKG2D required inverse orientation fusion of either of them. We hypothesized that the particularly located C-terminus of the NKG2D ectodomain should allow reengineering of the membrane anchoring from a native N-terminal to an artificial C-terminal linkage. Indeed, the resulting chimeric NKG2D receptor (chNKG2D) was functional and efficiently mediated ESFT cell death triggered by activated T cells. Notably, ESFT cells with even low NKG2D-L expression were killed by CD8(pos) and also CD4(pos) cells. Both, mRNA transfection and lentiviral transduction resulted in high level surface expression of chNKG2D. However, upon target-cell recognition receptor surface levels were maintained by tranfected RNA only during the first couple of hours after transfection. Later, target-cell contact resulted in strong and irreversible receptor down-modulation, whereas lentivirally mediated expression of chNKG2D remained constant under these conditions. Together, our study defines NKG2D-Ls as targets for a CAR-mediated T cell based immunotherapy of ESFT. A comparison of two different methods of gene transfer reveals strong differences in the susceptibility to ligand-induced receptor down-modulation with possible implications for the applicability of RNA transfection.


Subject(s)
Immunotherapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , RNA, Messenger/administration & dosage , Recombinant Fusion Proteins/metabolism , Sarcoma, Ewing/immunology , Sarcoma, Ewing/therapy , T-Lymphocytes/immunology , Apoptosis , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/transplantation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...