Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Front Immunol ; 10: 1962, 2019.
Article in English | MEDLINE | ID: mdl-31555260

ABSTRACT

Anti-tumor immunity is limited by a number of factors including the lack of fully activated T-cells, insufficient antigenic stimulation and the immune-suppressive tumor microenvironment. We addressed these hurdles by developing a novel class of immunoconjugates, Antibody-Targeted Pathogen-derived Peptides (ATPPs), which were designed to efficiently deliver viral T-cell epitopes to tumors with the aim of redirecting virus-specific memory T-cells against the tumor. ATPPs were generated through covalent binding of mature MHC class I peptides to antibodies specific for cell surface-expressed tumor antigens that mediate immunoconjugate internalization. By means of a cleavable linker, the peptides are released in the endosomal compartment, from which they are loaded into MHC class I without the need for further processing. Pulsing of tumor cells with ATPPs was found to sensitize these for recognition by virus-specific CD8+ T-cells with much greater efficiency than exogenous loading with free peptides. Systemic injection of ATPPs into tumor-bearing mice enhanced the recruitment of virus-specific T-cells into the tumor and, when combined with immune checkpoint blockade, suppressed tumor growth. Our data thereby demonstrate the potential of ATPPs as a means of kick-starting the immune response against "cold" tumors and increasing the efficacy of checkpoint inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/administration & dosage , Immunoconjugates/administration & dosage , Neoplasms/therapy , Peptides/administration & dosage , Animals , Cell Line, Tumor , Female , Herpesvirus 4, Human , Humans , Immunotherapy , Mice
2.
Mol Oncol ; 7(6): 1142-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055141

ABSTRACT

CUB-domain-containing-protein-1 (CDCP1) is an integral membrane protein whose expression is up-regulated in various cancer types. Although high CDCP1 expression has been correlated with poor prognosis in lung, breast, pancreas, and renal cancer, its functional role in tumor formation or progression is incompletely understood. So far it has remained unclear, whether CDCP1 is a useful target for antibody therapy of cancer and what could be a desired mode of action for a therapeutically useful antibody. To shed light on these questions, we have investigated the cellular effects of a therapeutic antibody candidate (RG7287). In focus formation assays, prolonged RG7287 treatment prevented the loss of contact inhibition caused by co-transformation of NIH3T3 cells with CDCP1 and Src. In a xenograft study, MCF7 cells stably overexpressing CDCP1 reached the predefined tumor volume faster than the parental MCF7 cells lacking endogenous CDCP1. This tumor growth advantage was abolished by RG7287 treatment. In vitro, RG7287 induced rapid tyrosine phosphorylation of CDCP1 by Src, which was accompanied by translocation of CDCP1 to a Triton X-100 insoluble fraction of the plasma membrane. Triggering these effects required bivalency of the antibody suggesting that it involves CDCP1 dimerization or clustering. However, this initial activation of CDCP1 was only transient and prolonged RG7287 treatment induced internalization and down-regulation of CDCP1 in different cancer cell lines. Antibody stimulated CDCP1 degradation required Src activity and was proteasome dependent. Also in three different xenograft models with endogenous CDCP1 expression RG7287 treatment resulted in significant tumor growth inhibition concomitant with substantially reduced CDCP1 levels as judged by immunohistochemistry and Western blotting. Thus, despite transiently activating CDCP1 signaling, the RG7287 antibody has a therapeutically useful mode of action.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antigens, CD , Antigens, Neoplasm , Cell Adhesion Molecules , Cell Membrane/metabolism , Membrane Glycoproteins , Neoplasm Proteins , Neoplasms, Experimental , Proteolysis/drug effects , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Membrane/pathology , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice , NIH 3T3 Cells , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 102(44): 16013-8, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16247014

ABSTRACT

Our understanding of pathways leading to antitumor immunity may depend on an undistorted knowledge of the primary antigenic targets of patients' autologous T cell responses. In the melanoma model derived from patient DT, we applied cryopreserved short-term autologous mixed lymphocyte-tumor cell cultures (MLTCs) in combination with an IFN-gamma enzyme-linked immunospot (ELISPOT) assay to cDNA expression screening. We identified three previously unknown peptides processed from melanosomal proteins tyrosinase (presented by HLA-A(*)2601 and -B(*)3801) and gp100 (presented by HLA-B(*)07021) and five neoantigens generated by somatic point mutations in the patient's melanoma. The mutations were found in the genes SIRT2, GPNMB, SNRP116, SNRPD1, and RBAF600. Peptides containing the mutated residues were presented by HLA-A(*)03011, -B(*)07021, and -B(*)3801. Mutation-induced functional impairment was so far demonstrated for SIRT2. Within MLTC responder populations that were independently expanded from the patient's peripheral blood lymphocytes of different years, T cells against mutated epitopes clearly predominated. These results document a high degree of individuality for the cellular antitumor response and support the need for individualizing the monitoring and therapeutic approaches to the primary targets of the autologous T cell response, which may finally lead to a more effective cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/genetics , Melanoma/immunology , Point Mutation , T-Lymphocytes/immunology , Adult , Antigen Presentation , Antigens, Neoplasm/immunology , Base Sequence , Coculture Techniques , Epitopes, T-Lymphocyte , Female , HLA Antigens/immunology , Humans , Interferon-gamma/analysis , Melanoma/pathology , Molecular Sequence Data , T-Lymphocytes/cytology , Tumor Cells, Cultured
4.
Blood ; 104(8): 2591-9, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15231579

ABSTRACT

Allogeneic hematopoietic stem cell transplantation can induce considerable tumor remissions in metastatic renal-cell carcinoma (RCC) patients. The precise effector mechanisms mediating these graft-versus-tumor reactions are unknown. We studied RCC-directed CD8(+) T-cell responses in blood lymphocytes of healthy individuals matched with established RCC cell lines for HLA-class I. In 21 of 22 allogeneic mixed lymphocyte/tumor-cell cultures (MLTCs), RCC-reactive cytotoxic T-lymphocytes (CTLs) were readily obtained. From MLTCs, 121 CD8(+) CTL clones with memory phenotype were isolated. Their anti-RCC reactivity was restricted by multiple classical HLA-Ia molecules, in particular by HLA-A2, -A3, -B7, -B44, -Cw7, and by a nonclassical HLA-Ib determinant. Extensive cross-reactivity analyses on a broad target panel identified CTLs that recognize antigens with expression restricted to renal tissue or to renal and colon tumors. Other CTLs were directed against antigens with broader tissue distribution being expressed in various epithelial and nonepithelial tumors or, additionally, in hematopoietic cells. With microcapillary liquid chromatography and matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF)/TOF mass spectrometry, we identified the HLA-A*0301-associated nonpolymorphic peptide KLPNSVLGR encoded by the ubiquitously expressed Eps15 homology domain-containing 2 gene as a CTL target. Defining human RCC antigens recognized by alloreactive CTLs may allow to improve the specificity and efficiency of allogeneic cell therapy (eg, specific donor-lymphocyte infusions or vaccination) in metastatic RCC patients.


Subject(s)
Carcinoma, Renal Cell/immunology , Histocompatibility Antigens Class I/immunology , Mitogen-Activated Protein Kinases/immunology , Mitogen-Activated Protein Kinases/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous , Amino Acid Sequence , Antigens, Neoplasm , CD4-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/metabolism , Cell Separation , Colonic Neoplasms/immunology , Cross Reactions , Cytotoxicity, Immunologic , Epithelium/immunology , Epitopes/chemistry , Epitopes/immunology , Flow Cytometry , Genotype , Health , Hematopoietic Stem Cells/immunology , Histocompatibility Antigens Class I/genetics , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Donors
SELECTION OF CITATIONS
SEARCH DETAIL
...