Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
Genes Immun ; 15(2): 115-25, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24401767

ABSTRACT

Disturbed expression of microRNAs (miRNAs) in regulatory T cells (Tregs) leads to development of autoimmunity in experimental mouse models. However, the miRNA expression signature characterizing Tregs of autoimmune diseases, such as rheumatoid arthritis (RA) has not been determined yet. In this study, we have used a microarray approach to comprehensively analyze miRNA expression signatures of both naive Tregs (CD4+CD45RO-CD25++) and memory Tregs (CD4+CD45RO+CD25+++), as well as conventional naive (CD4+CD45RO-CD25-) and memory (CD4+CD45RO+CD25-) T cells (Tconvs) derived from peripheral blood of RA patients and matched healthy controls. Differential expression of selected miRNAs was validated by TaqMan-based quantitative reverse transcription-PCR. We found a positive correlation between increased expression of miR-451 in T cells of RA patients and disease activity score (DAS28), erythrocyte sedimentation rate levels and serum levels of interleukin-6. Moreover, we found characteristic, disease- and treatment-independent, global miRNA expression signatures defining naive Tregs, memory Tregs, naive Tconvs and memory Tconvs. The analysis allowed us to define miRNAs characteristic for a general naive phenotype (for example, miR-92a) and a general memory phenotype (for example, miR-21, miR-155). Importantly, the analysis allowed us to define miRNAs that are specifically expressed in both naive and memory Tregs, defining as such miRNA signature characterizing the Treg phenotype (that is, miR-146a, miR-3162, miR-1202, miR-1246 and miR-4281).


Subject(s)
Arthritis, Rheumatoid/genetics , MicroRNAs/genetics , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Blood Sedimentation , CD4 Antigens/genetics , Female , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-6/blood , Leukocyte Common Antigens/genetics , Male , MicroRNAs/biosynthesis , Middle Aged , Oligonucleotide Array Sequence Analysis , Synovial Fluid/cytology , Synovial Fluid/immunology
3.
Oncogene ; 26(26): 3769-76, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17173072

ABSTRACT

BIC is a primary microRNA (pri-miR-155) that can be processed to mature miR-155. In this study, we show the crucial involvement of protein kinase C (PKC) and nuclear factor-kappaB (NF-kappaB) in the regulation of BIC expression upon B-cell receptor triggering. Surprisingly, Northern blot analysis did not reveal any miR-155 expression upon induction of BIC expression in the Burkitt lymphoma-derived Ramos cell line, whereas other microRNAs were clearly detectable. Ectopic expression of BIC in Ramos and HEK293 cells resulted in miR-155 expression in HEK293, but not in Ramos cells, suggesting a specific block of BIC to miR-155 processing in Ramos. In line with the results obtained with Ramos, lack of miR-155 expression after induction of BIC expression was also observed in other Burkitt lymphoma cell lines, indicating a generic and specific blockade in the processing of BIC in Burkitt lymphoma. In contrast, induction of BIC expression in normal tonsillar B cells resulted in very high levels of miR-155 expression and induction of BIC expression in Hodgkin's lymphoma cell lines. It also resulted in elevated levels of miR-155. Our data provide evidence for two levels of regulation for mature miR-155 expression: one at the transcriptional level involving PKC and NF-kappaB, and one at the processing level. Burkitt lymphoma cells not only express low levels of BIC, but also prevent processing of BIC via an, as yet, unknown mechanism.


Subject(s)
Burkitt Lymphoma/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA Processing, Post-Transcriptional , Blotting, Northern , Cell Line, Tumor , Humans , NF-kappa B/metabolism , Protein Kinase C/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
4.
Leukemia ; 20(11): 1931-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16990772

ABSTRACT

Over the past few years, it has become evident that microRNAs (miRNAs) play an important regulatory role in various biological processes. Much effort has been put into the elucidation of their biogenesis, and this has led to the general concept that a number of key regulators are shared with the processing machinery of small interfering RNAs. Despite the recognition that several miRNAs play crucial roles in normal development and in diseases, little is known about their exact molecular function and the identity of their target genes. In this review, we report on the biological relevance of miRNAs for the differentiation of normal hematopoietic cells and on the contribution of deregulated miRNA expression in their malignant counterparts.


Subject(s)
Gene Expression Regulation, Leukemic , Hematologic Neoplasms/genetics , Hematologic Neoplasms/physiopathology , Hematopoiesis/genetics , MicroRNAs/physiology , Animals , Humans
5.
Crit Rev Oncol Hematol ; 40(1): 53-76, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11578916

ABSTRACT

A wide range of strategies in cancer immunotherapy has been developed in the last decade, some of which are currently being used in clinical settings. The development of these immunotherapeutical strategies has been facilitated by the generation of relevant transgenic animal models. Since the different strategies in experimental immunotherapy of cancer each aim to activate different immune system components, a variety of transgenic animals have been generated either expressing tumor associated, HLA, oncogenic or immune effector cell molecule proteins. This review aims to discuss the existing transgenic mouse models generated to study and develop cancer immunotherapy strategies and the variable results obtained. The potential of the various transgenic animal models regarding the development of anti-cancer immunotherapeutical strategies is evaluated.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Histocompatibility Antigens/genetics , Humans , Mice , Mice, Transgenic , Neoplasms, Experimental
6.
J Biol Chem ; 276(48): 44848-55, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11502750

ABSTRACT

The search for potential targets for ceramide action led to the identification of ceramide-activated protein phosphatases (CAPP). To date, two serine/threonine protein phosphatases, protein phosphatase 2A (PP2A) and protein phosphatase 1 (PP1), have been demonstrated to function as ceramide-activated protein phosphatases. In this study, we show that treatment with either anti-FAS IgM (CH-11) (150 ng/ml) or exogenous d-(e)-C(6-)ceramide (20 microm) induces the dephosphorylation of the PP1 substrates, serine/arginine-rich (SR) proteins, in Jurkat acute leukemia T-cells. The serine/threonine protein phosphatase inhibitor, calyculin A, but not the PP2A-specific inhibitor, okadaic acid, inhibited both FAS- and ceramide-induced dephosphorylation of SR proteins. Anti-FAS IgM treatment of Jurkat cells led to a significant increase in levels of endogenous ceramide beginning at 2 h with a maximal increase of 10-fold after 7 h. A 2-h pretreatment of Jurkat cells with fumonisin B(1) (100 microm), a specific inhibitor of CoA-dependent ceramide synthase, blocked 80% of the ceramide generated and completely inhibited the dephosphorylation of SR proteins in response to anti-FAS IgM. Moreover, pretreatment of Jurkat cells with myriocin, a specific inhibitor of serine-palmitoyl transferase (the first step in de novo synthesis of ceramide), also blocked FAS-induced SR protein dephosphorylation, thus demonstrating a role for de novo ceramide. These results were further supported using A549 lung adenocarcinoma cells treated with d-(e)-C(6-)ceramide. Dephosphorylation of SR proteins was inhibited by fumonisin B(1) and by overexpression of glucosylceramide synthase; again implicating endogenous ceramide generated de novo in regulating the dephosphorylation of SR proteins in response to FAS activation. These results establish a specific intracellular pathway involving both de novo ceramide generation and activation of PP1 to mediate the effects of FAS activation on SR proteins.


Subject(s)
Ceramides/biosynthesis , Fumonisins , Phosphoprotein Phosphatases/metabolism , Sarcoplasmic Reticulum/metabolism , fas Receptor/metabolism , Blotting, Western , Carboxylic Acids/pharmacology , Cell Line , Diacylglycerol Kinase/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Jurkat Cells , Palmitic Acid/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Phosphatase 1 , Protein Phosphatase 2 , Serine/metabolism , Sphingolipids/metabolism , Threonine/metabolism , Time Factors
7.
Cancer Res ; 61(10): 4105-11, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358833

ABSTRACT

The human pancarcinoma-associated epithelial glycoprotein-2 (EGP-2), a M(r) 38,000 transmembrane antigen also known as 17-1A or Ep-CAM, is commonly used for targeted immunotherapy of carcinomas because it is strongly expressed by most carcinomas. EGP-2 is, however, also expressed in most normal epithelia. To evaluate anti-EGP-2-directed treatment-associated effects on tumors and on EGP-2-positive normal tissue, we generated EGP-2-expressing transgenic mice. A 55-kb DNA fragment consisting of the 14-kb genomic coding sequence of the human EGP-2 gene with approximately 10-kb-upstream and approximately 31-kb-downstream sequences was isolated and used to direct EGP-2 expression in an epithelium-specific manner. In the EGP-2 transgenic mice, EGP-2 appeared to be specifically expressed in all of those epithelial tissues that also express EGP-2 in humans, whereas all of the other tissues were negative. The specific in vivo localization of the i.v. administered anti-EGP-2 monoclonal antibody MOC31 was studied in EGP-2-positive and -negative tumors induced s.c. in this EGP-2 transgenic mouse model. Immunohistochemical analysis showed specific localization of MOC31 in the EGP-2-positive tumors but not in the EGP-2-negative tumors. No anti-EGP-2 monoclonal antibody localization was observed in any of the EGP-2-positive normal mouse tissues, which indicated a limited in vivo accessibility. In conclusion, an EGP-2 transgenic mouse model has been generated that expresses the EGP-2 antigen as in humans and, therefore, can serve as a model to evaluate the efficacy and safety of a variety of anti-EGP-2-based immunotherapeutic modalities in both tumors and normal tissue.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Immunotherapy/methods , Melanoma, Experimental/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigens, Neoplasm/biosynthesis , Cell Adhesion Molecules/biosynthesis , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Female , Humans , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic
8.
Br J Cancer ; 84(8): 1115-21, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11308263

ABSTRACT

This paper describes a bi-specific antibody, which was called BIS20x3. It retargets CD3varepsilon-positive cells (T-cells) to CD20-positive cells and was obtained by hybrid-hybridoma fusion. BIS20x3 could be isolated readily from quadroma culture supernatant and retained all the signalling characteristics associated with both of its chains. Cross-linking of BIS20x3 on Ramos cells leads to DNA fragmentation percentages similar to those obtained after Rituximab-cross-linking. Cross-linking of BIS20x3 on T-cells using cross-linking F(ab')2-fragments induced T-cell activation. Indirect cross-linking of T-cell-bound BIS20x3 via Ramos cells hyper-activated the T-cells. Furthermore, it was demonstrated that BIS20x3 effectively re-targets T-cells to B-cells, leading to high B-cell cytotoxicity. The results presented in this paper show that BIS20x3 is fully functional in retargeting T-cells to B-cells and suggest that B-cell lymphomas may represent ideal targets for T-cell retargeting bi-specific antibodies, because the retargeted T-cell is maximally stimulated in the presence of B-cells. Additionally, since B-cells may up-regulate CD95/ Fas expression upon binding of CD20-directed antibodies, B-cells will become even more sensitive for T-cell mediated killing via CD95L/ Fas L, and therefore supports the intention to use T-cell retargeting bi-specific antibodies recognizing CD20 on B-cell malignancies as a treatment modality for these diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD20/immunology , B-Lymphocytes/immunology , CD3 Complex , T-Lymphocytes/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , B-Lymphocytes/cytology , Cell Fusion , Cell Line , DNA/drug effects , DNA/metabolism , Dose-Response Relationship, Drug , Humans , Hybridomas/cytology , Hybridomas/immunology , Jurkat Cells , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/cytology , Tumor Cells, Cultured
9.
J Biol Chem ; 276(17): 13606-14, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11278517

ABSTRACT

B-cells, triggered via their surface B-cell receptor (BcR), start an apoptotic program known as activation-induced cell death (AICD), and it is widely believed that this phenomenon plays a role in the restriction and focusing of the immune response. Although both ceramide and caspases have been proposed to be involved in AICD, the contribution of either and the exact molecular events through which AICD commences are still unknown. Here we show that in Ramos B-cells, BcR-triggered cell death is associated with an early rise of C16 ceramide that derives from activation of the de novo pathway, as demonstrated using a specific inhibitor of ceramide synthase, fumonisin B1 (FB1), and using pulse labeling with the metabolic sphingolipid precursor, palmitate. There was no evidence for activation of sphingomyelinases or hydrolysis of sphingomyelin. Importantly, FB1 inhibited several specific apoptotic hallmarks such as poly(A)DP-ribose polymerase cleavage and DNA fragmentation. Electron microscopy revealed morphological evidence of mitochondrial damage, suggesting the involvement of mitochondria in BcR-triggered apoptosis, and this was inhibited by FB1. Moreover, a loss of mitochondrial membrane potential was observed in Ramos cells after BcR cross-linking, which was inhibited by the addition of FB1. Interestingly, benzyloxycarbonyl-Val-Ala-dl-Asp, a broad spectrum caspase inhibitor did not inhibit BcR-induced mitochondrial membrane permeability transition but did block DNA fragmentation. These results suggest a crucial role for de novo generated C16 ceramide in the execution of AICD, and they further suggest an ordered and more specific sequence of biochemical events in which de novo generated C16 ceramide is involved in mitochondrial damage resulting in a downstream activation of caspases and apoptosis.


Subject(s)
Apoptosis , B-Lymphocytes/metabolism , Ceramides/metabolism , Cross-Linking Reagents/pharmacology , Fumonisins , Mitochondria/metabolism , Receptors, Antigen, B-Cell/metabolism , Transcription Factors , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Carboxylic Acids/pharmacology , Chromatography, High Pressure Liquid , DNA Fragmentation , DNA-Binding Proteins/metabolism , Diacylglycerol Kinase/metabolism , Dose-Response Relationship, Drug , Humans , Mass Spectrometry , Membrane Potentials , Microscopy, Electron , Mitochondria/ultrastructure , Oxidoreductases/antagonists & inhibitors , Palmitates/metabolism , Time Factors
10.
Clin Cancer Res ; 6(6): 2521-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10873108

ABSTRACT

Peripheral blood stem cell (PBSC) support in breast cancer patients allows high-dose chemotherapy, but tumor cell contamination of the PBSCs is a potential source of relapse. Specific carcinoma cell killing can be obtained by retargeting activated T cells with bispecific antibody BIS-1, directed against epithelial glycoprotein-2 and CD3. To purge epithelial tumor cells from the PBSCs of breast cancer patients, activation of T cells in PBSCs and T-cell retargeting by BIS-1 was studied. PBSCs, obtained by leukapheresis after chemotherapy and recombinant human granulocyte colony-stimulating factor, were cultured in the presence of PBS, interleukin-2, OKT3, or interleukin-2/OKT3 for induction of T-cell activation. Subsequently, lysis of epithelial tumor cell lines by activated T cells of PBSCs in the presence or absence of BIS-1 was assessed with the 51Cr-release assay or immunocytochemical staining. The effect on PBSC hematopoietic colony formation (HCF) was evaluated by the granulocyte macrophage colony-stimulating units assay. Prior to activation, PBSCs from breast cancer patients contained higher levels of CD8+ T cells than peripheral blood from healthy volunteers (P < 0.05). The potential of PBSCs to sustain tumor cell lysis was increased after all prior activations and was further enhanced by BIS-1. Maximal BIS-1 effect was observed after OKT3 activation of PBSCs for 72 h (P < 0.0005), inducing a >3 log depletion of tumor cells. HCF was not affected by prior OKT3 activation and/or BIS-1. In conclusion, specific tumor cell lysis by PBSCs can be obtained in vitro by OKT3 activation and BIS-1 retargeting of T cells, without affecting HCF. At present, studies are evaluating this format for future clinical application.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Lymphocyte Activation , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/therapy , Stem Cells/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Antibodies/metabolism , Breast Neoplasms/blood , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/metabolism , Chromium Radioisotopes/metabolism , Female , Flow Cytometry , Glycoproteins/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunohistochemistry , Immunotherapy/methods , Interleukin-2/pharmacology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Neoplasms, Glandular and Epithelial/blood , Stem Cells/immunology , Tumor Cells, Cultured
11.
J Immunol Methods ; 237(1-2): 131-45, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10725458

ABSTRACT

A versatile expression vector is described for the rapid construction and evaluation of bispecific scFvs and scFv-based fusion proteins. An important feature of this vector is the presence of two multiple cloning sites (MCS) separated by an in frame linker sequence. The first MCS was specifically designed to contain unique SfiI and NotI restriction enzyme sites that can be used for directional and in frame insertion of scFvs (or potentially any molecule) selected from established phage-display systems. Using this new vector, a functional bs-(scFv)(2) (2C11-MOC31) was constructed for retargeted T-cell cytotoxicity towards EGP2 positive tumor cells. The vector was also used for grafting of a number of promising biological effector principles onto scFv MOC31, including the prodrug converting enzyme cytosine deaminase, the anti-angiogenic factor angiostatin, and the thrombogenic molecule tissue factor. We aimed at producing biologically active fusion proteins by directing them through the endoplasmic reticulum-based protein folding machinery of eukaryotic cells (COS-7) using a kappa light chain leader, thereby taking advantage of the associated quality control mechanisms that allow only fully folded and processed fusion proteins to be secreted into the medium. Supernatants derived from fusion protein transfected COS-7 cells, which were transiently transfected at low transfection rates, were directly assayed for the biological and/or targeting activity of the excreted fusion proteins without any prior purification steps. This procedure might help to identify those fusion proteins that have favourable characteristics like stability and biological activity in the presence of serum and at low protein concentrations. Targeted delivery of all effector principles was subsequently assessed in an in vitro model system. The method we devised is both rapid and versatile and can be useful to construct and identify series of new chimeric proteins with enhanced therapeutic potential in human cancer therapy.


Subject(s)
Genetic Vectors , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/isolation & purification , Angiostatins , Animals , Base Sequence , COS Cells , Cytosine Deaminase , Cytotoxicity, Immunologic , DNA Primers/genetics , Gene Expression , Humans , Immunoglobulin Fragments/biosynthesis , In Vitro Techniques , Mice , Molecular Sequence Data , Nucleoside Deaminases/biosynthesis , Nucleoside Deaminases/genetics , Nucleoside Deaminases/isolation & purification , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Plasmids/genetics , Plasminogen/biosynthesis , Plasminogen/genetics , Plasminogen/isolation & purification , Protein Folding , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , T-Lymphocytes, Cytotoxic/immunology , Thromboplastin/biosynthesis , Thromboplastin/genetics , Thromboplastin/isolation & purification , Transfection , Tumor Cells, Cultured
12.
Br J Cancer ; 82(2): 472-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10646907

ABSTRACT

Bispecific antibody (BsMAb) BIS-1 has been developed to redirect the cytolytic activity of cytotoxic T lymphocytes (CTL) to epithelial glycoprotein-2 (EGP-2) expressing tumour cells. Intravenous administration of BIS-1 F(ab')2 to carcinoma patients in a phase I/II clinical trial, caused immunomodulation as demonstrated by a rapid lymphopenia prior to a rise in plasma tumour necrosis factor-alpha and interferon-gamma levels. Yet, no lymphocyte accumulation in the tumour tissue and no anti-tumour effect could be observed. These data suggest a BsMAb-induced lymphocyte adhesion to blood vessel walls and/or generalized redistribution of the lymphocytes into tissues. In this study, we describe the effects of BIS-1 F(ab')2 binding to peripheral blood mononuclear cells (PBMC) on their capacity to interact with resting endothelial cells in vitro. Resting and pre-activated PBMC exhibited a significant increase in adhesive interaction with endothelial cells when preincubated with BIS-1 F(ab')2, followed by an increase in transendothelial migration (tem). Binding of BIS-1 F(ab')2 to PBMC affected the expression of a number of adhesion molecules involved in lymphocyte adhesion/migration. Furthermore, PBMC preincubated with BIS-1 F(ab')2 induced the expression of endothelial cell adhesion molecules E-selectin, VCAM-1 and ICAM-1 during adhesion/tem. These phenomena were related to the CD3 recognizing antibody fragment of the BsMAb and dependent on lymphocyte-endothelial cell contact. Possibly, in patients, the BIS-1 F(ab')2 infusion induced lymphopenia is a result of generalized activation of endothelial cells, leading to the formation of a temporary sink for lymphocytes. This process may distract the lymphocytes from homing to the tumour cells, and hence prevent the occurrence of BIS-1 F(ab')2 - CTL-mediated tumour cell lysis.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Cell Adhesion/immunology , Lymphocytes/immunology , Cell Communication/immunology , Cell Movement , Endothelium/cytology , Endothelium/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunotherapy , In Vitro Techniques , Intercellular Adhesion Molecule-1/immunology , Lymphopenia/immunology , Vascular Cell Adhesion Molecule-1/immunology
13.
J Control Release ; 64(1-3): 229-39, 2000 Feb 14.
Article in English | MEDLINE | ID: mdl-10640660

ABSTRACT

To overcome dose limiting toxicities and to increase efficacy of immunotherapy of cancer, a number of strategies are under development for selectively redirecting effector cells/molecules towards tumor cells. Many of these strategies exploit the specificity of tumor associated antigen recognition by monoclonal antibodies. Using either hybridoma fusion, chemical derivatization or molecular biology technology, antibodies with dual specificity can be constructed. These so called biospecific antibodies (BsAbs) have been used to redirect the cytolytic activity of a variety of immune effector cells such as cytotoxic T lymphocytes, natural killer cells, neutrophils and monocytes/macrophages to tumor cells. Local administration of BsAbs, either alone or in combination with autologous effector cells, is highly effective in eradicating tumor cells. In contrast, systemic application of BsAb at present is only suitable for adjuvant treatment of minimal residual disease due to poor tumor cell accessibility. As an alternative, angiogenesis related determinants on tumor blood vessels can be exploited for the selective delivery of effector cells/molecules apart from being used to inhibit angiogenesis. Important advantages of this strategy is that the endothelial cell associated target epitope(s) are easy accessible. The dependence of tumor growth on the tumor's blood supply also renders tumor endothelial cells an attractive target for therapy. Although still in its infancy, attacking the tumor's blood supply for example by delivering coagulation factors or toxins, or by BsAb directed immunotherapies holds great promise for antineoplastic therapy.


Subject(s)
Antibodies, Bispecific/therapeutic use , Drug Delivery Systems/methods , Immunotherapy/methods , Neoplasms/therapy , Neovascularization, Pathologic/prevention & control , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/administration & dosage , Blood Vessels/drug effects , Clinical Trials as Topic , Epitopes/drug effects
14.
Cancer Immunol Immunother ; 48(6): 303-11, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10473805

ABSTRACT

The human pancarcinoma-associated epithelial glycoprotein-2 (EGP-2), also known as 17-1A or Ep-CAM, is a 38-kDa transmembrane antigen, commonly used for targeted immunotherapy of carcinomas. Although strongly expressed by most carcinomas, EGP-2 is also expressed in most simple epithelia. To evaluate treatment-associated effects and side-effects on tumor and normal tissue respectively, we generated an EGP-2-expressing transgenic Wistar rat. To express the cDNA of the EGP-2 in an epithelium-specific manner, the 5' and 3' distal flanking regions of the human keratin 18 (K18) gene were used. EGP-2 protein expression was observed in the liver and pancreas, whereas EGP-2 mRNA could also be detected in lung, intestine, stomach and kidney tissues. In this rat, EGP-2-positive tumors can be induced by injecting a rat-derived carcinoma cell line transfected with the GA733-2 cDNA encoding EGP-2. Transgenic rats were used to study specific in vivo localization of an i.v. anti-EGP-2 monoclonal antibody, MOC31, applied i.v. Immunohistochemical analyses showed the specific localization of MOC31 in s.c. induced EGP-2-positive tumors, as well as in the liver. In contrast, in EGP-2-transgenic rats, MOC31 did not bind to EGP-2-negative tumors, the pancreas, or other normal tissues in vivo. In conclusion, an EGP-2-transgenic rat model has been generated that serves as a model to evaluate the efficacy and safety of a variety of anti-EGP-2-based immunotherapeutic modalities.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm/genetics , Carcinoma/therapy , Cell Adhesion Molecules/genetics , Immunization, Passive , Animals , Animals, Genetically Modified , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Carcinoma/immunology , Carcinoma/pathology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/immunology , DNA, Complementary/genetics , Epithelial Cell Adhesion Molecule , Epithelial Cells/metabolism , Female , Humans , Immunoenzyme Techniques , Keratins/genetics , Male , Neoplasm Transplantation , Organ Specificity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Transfection , Tumor Cells, Cultured , Viscera/metabolism
15.
Cytometry ; 32(1): 37-43, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9581622

ABSTRACT

In this report, we describe a new and simple method for flow cytometric quantitation of lymphocyte numbers in lymphocyte-endothelial adhesion/ transendothelial migration assays. The method exploits fluorescent flow cytometer alignment beads as a counting reference. Known amounts of beads are added to samples with unknown lymphocyte numbers. Lymphocytes and a preset number of fluorescent beads are simultaneously analyzed. The total number of cells present in the sample can be subsequently calculated from the fixed ratio of added to analyzed fluorescent beads. Using this fluorescent-beads-based flow cytometric cell counting of lymphocyte numbers in adhesion/migration assays, labeling of cells and other time-consuming calibration procedures are not required and analysis time is short. Furthermore, we demonstrate that this cell counting method can be combined with concurrent single- or double-label fluorescence flow cytometric phenotyping of adherent and migrated lymphocytes. The method was applied to the in vitro study of the effects of lymphocyte activation status and binding of bispecific antibody (directed against CD3 x tumor cell-associated antigen) on lymphocyte adhesion and transendothelial migration.


Subject(s)
Cell Adhesion/immunology , Cell Movement/immunology , Endothelium, Vascular/immunology , Flow Cytometry/methods , Lymphocytes/cytology , Antibodies, Monoclonal , Antigens, Neoplasm/analysis , CD3 Complex/analysis , Cell Adhesion Molecules/analysis , Cell Count/methods , Cell Separation , Cells, Cultured , Epithelial Cell Adhesion Molecule , Humans , Immunoglobulin Fab Fragments , Lymphocyte Activation , Microspheres , Phenotype , Reproducibility of Results , Umbilical Veins
16.
Int J Cancer ; 76(2): 232-9, 1998 Apr 13.
Article in English | MEDLINE | ID: mdl-9537586

ABSTRACT

We describe the construction of a recombinant bispecific antibody fragment in the diabody format with specificity for both the well-established human pancarcinoma associated target antigen EGP2 (epithelial glycoprotein 2, also known as the CO17-1A antigen or KSA) and the CD3epsilon chain of human TCR/CD3 complex. The murine anti-EGP2 (MOC31) single chain variable fragment (scFv) and the humanized anti-CD3 (Ucht1v9) scFv were cast into a diabody format (designated Dia5v9) using a short 5 amino acid Gly-Ser linker between immunoglobulin heavy-chain and light-chain variable domains. Purification of the poly-histidine tagged Dia5v9 was achieved from extracts of protease deficient Escherichia coli by IMAC chromatography. The Dia5v9 diabody showed strong binding to both EGP2 and CD3 in transfected cells. The in vitro efficacy of Dia5v9 in mediating tumor cell lysis by interleukin-2 activated human T cells appeared to be similar to that of the hybrid-hybridoma-derived BsF(ab')2 Bis1 (anti-EGP2/anti-CD3) in a standard 4-hr 51Cr-release assay. This small and partially humanized recombinant bispecific antibody fragment may be valuable for T-cell-based immunotherapeutical treatment protocols, retargeting activated peripheral blood T lymphocytes to lyse various human carcinomas in vivo.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/pharmacology , Immunoglobulin Fragments/biosynthesis , Immunoglobulin Fragments/pharmacology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/metabolism , Antibody Specificity , Antigens, Neoplasm/immunology , COS Cells/metabolism , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Humans , Immunization, Passive , Immunoglobulin Fragments/metabolism , Lymphocyte Activation/immunology , Neoplasms/therapy , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sodium Dodecyl Sulfate , T-Lymphocytes, Cytotoxic/immunology
17.
Cancer Immunol Immunother ; 45(3-4): 203-6, 1997.
Article in English | MEDLINE | ID: mdl-9435874

ABSTRACT

The bispecific monoclonal antibody (bsAb) BIS-1 combines a monoclonal-antibody(mAb)-defined specificity for the CD3 complex, as present on all T lymphocytes, with a mAb-defined specificity for the pancarcinoma/epithelium associated glycoprotein EGP-2. In vitro studies indicate that BIS-1 can direct T lymphocytes to kill EGP-2-positive tumour target cells. T cell pre-activation is necessary for this activity and can be obtained either via incubation of isolated peripheral blood mononuclear cells with CD3 mAb, followed by short culturing in recombinant interleukin-2-containing medium, or via costimulation with CD5- and CD28-based bsAb. Clinical application of BIS-1 was started in a pilot study in which carcinoma patients suffering from malignant ascites or intrapleural effusion were treated. In this study, ex vivo activated autologous lymphocytes were applied locally, i.e. intraperitoneally or intrapleurally, in the presence of BIS-1. Local inflammation and antitumour activity were observed, whereas no or only minor systemic toxicity was seen in these patients. Intravenous administration of BIS-1 F(ab')2 in combination with subcutaneously given recombinant interleukin-2 (i.v. bsAb/rIL-2 treatment) induced transient but considerable toxicity including peripheral vasoconstriction, dyspnoea and fever with a maximal tolerated dose of 5-8 micrograms/kg. High plasma concentrations of the inflammatory cytokines tumor necrosis factor alpha and interferon gamma were observed at this dose. Whereas bsAb-dictated antitumour activity could be demonstrated to be present in blood samples of these patients in an in vitro assay, no clear clinical responses were observed. In a rat model it was found that i.v. bsAb/rIL-2 treatment of EGP-2-positive tumours was effective when a low systemic tumour burden was present, suggesting that systemic bsAb/rIL-2 treatment might be effective in situations of minimal residual disease.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Carcinoma, Renal Cell/therapy , Carcinoma, Squamous Cell/therapy , Cell Adhesion Molecules/immunology , Immunoglobulin Fab Fragments/therapeutic use , Kidney Neoplasms/therapy , Lung Neoplasms/therapy , Aged , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/secondary , Epithelial Cell Adhesion Molecule , Female , Humans , Immunoglobulin Fab Fragments/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Activation/immunology , Male , Middle Aged , Rats , T-Lymphocytes/immunology
18.
Br J Cancer ; 73(6): 721-7, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8611371

ABSTRACT

In this report we describe the role of apoptosis in the process of tumour cell killing by bispecific monoclonal antibody (BsMAb)-redirected cytolytic T cells. The BsMAb used, BIS-1, has dual specificity for the CD3 complex on T cells and the pancarcinoma-associated 38 kDa transmembrane antigen EGP-2. BIS-1 allows activated T cells to specifically recognise and kill EGP-2-positive but not EGP-2-negative target cells. An assay was developed to quantify apoptosis in cells by separation of 3H-thymidine-labelled low-molecular, i.e. fragmented, from high-molecular, i.e. non-fragmented DNA. The presence of low molecular weight DNA was measured both within the target cells and in the cell-free supernatant. After exposure to BIS-1-redirected, -activated T cells, apoptosis was observed in EGP-2-positive target cells but not in EGP-2-negative target cells. Also no DNA fragmentation proved to be induced in the activated effector cells during assay. The degree of EGP-2-positive target DNA fragmentation depended on the concentration of BsMAb, the E/T ratio and the incubation time. Using a low E/T ratio (1/1), DNA fragmentation in and 51Cr release from target cells showed similar characteristics and kinetics. At higher E/T ratio (20/1), the 51Cr release from the target cells increased to a greater extent than the percentage fragmented target cell DNA. Inhibitors of DNA fragmentation added to the cytotoxicity assay inhibited not only DNA fragmentation, but also the release of chromium-51 from the target cells, suggesting that apoptosis and cell lysis are closely related in BsMAb-mediated cell killing.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Apoptosis/physiology , T-Lymphocytes, Cytotoxic/immunology , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Apoptosis/drug effects , DNA/metabolism , DNA Damage , Humans , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism
19.
Cancer Res ; 55(19): 4409-15, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7545540

ABSTRACT

Induction of T-cell activation requires multiple signals provided by cell surface receptor interactions and/or cytokines. T-cell stimulation via the T-cell receptor/CD3 complex provides an important initial activation event which, when combined with the proper costimulatory signals, results in an activated effector T cell. In this report, we have investigated the effectiveness of epithelial glycoprotein-2- (EGP-2) positive tumor target cells to induce specific T-cell stimulation via CD3, CD5, and CD28 using various combinations of bispecific monoclonal antibodies (BsMab) directed against CD3, CD5, or CD28 on the one hand and the pancarcinoma-associated antigen EGP-2 on the other. Induction of T-cell activation was investigated by assessment of CD69 expression, induction of proliferation, and acquirement of cytolytic potential. EGP-2-specific induction of T-cell activation was observed using combinations of BsMab which simultaneous ligated CD3/CD5, CD3/CD28, or CD3/CD5/CD28 with EGP-2. Activation with CD3-, CD5-, or CD28-based BsMab alone did not result in significant induction of T-cell activation in the presence or absence of EGP-2-positive target cells. Simultaneous ligation via CD5/CD28 resulted in partial T-cell activation, including CD69 up-regulation and increased cytolytic activity. Stimulation via CD3 and CD5 or CD28 could be further increased by the addition of exogenously added recombinant Interleukin 2. In contrast, T-cell activation by simultaneous ligation of CD3/CD5/CD28 could not be further augmented by addition of exogenous interleukin 2, indicating that T-cell activation via the combination of CD3, CD5, and CD28 results in complete T-cell activation. Our results show that rapid and target cell-specific induction of T cells is possible using combinations of BsMab directed against different costimulatory molecules. Simultaneous costimulation via CD3/CD5/CD28 results in the most complete activation of T cells.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, CD/physiology , CD28 Antigens/physiology , Lymphocyte Activation , T-Lymphocytes/immunology , Antigens, Neoplasm/analysis , Antigens, Neoplasm/immunology , CD3 Complex/physiology , CD5 Antigens , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/immunology , Cytotoxicity, Immunologic , Epithelial Cell Adhesion Molecule , Humans , Interleukin-2/pharmacology , Tumor Cells, Cultured
20.
J Hematother ; 4(5): 409-14, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8581377

ABSTRACT

Immunobiologic parameters measured during a phase I trial of intravenously (i.v.) administered bispecific monoclonal antibodies (BsmAb) in renal cell carcinoma (RCC) patients are described. The BsmAb used, BIS-1, is reactive with a pancarcinoma-associated 38 kDa transmembrane glycoprotein, EGP-2, as well with the CD3 complex. Patients received during a 2 h i.v. infusion F(ab')2 fragments of BIS-1 at doses of 1, 3, or 5 micrograms/kg body weight during concomitantly applied subcutaneous (s.c.) IL-2 treatment. Acute but transient BIS-1 F(ab')2-related toxicity was observed at the 3 and 5 micrograms/kg dose level, and the maximum tolerated dose (MTD) was set at 5 micrograms/kg. A dose-dependent binding of BIS-1 F(ab')2 to circulating T lymphocytes was found. The in vivo occupancy of CD3 molecules on T lymphocytes was highest at teh end of the infusion period and then rapidly decreased, as shown by flow cytometry. A much slower decrease of BIS-1 F(ab')2 binding was observed in vitro, suggesting migration of BIS-1 F(ab')2-loaded T lymphocytes from the circulation. A strong but transitory leukopenia was observed, in which LFA-1 alpha bright, CD3/CD8 double positive T cells left the circulation preferentially. This phenomenon was most likely induced by elevated TNF-alpha and IFN-gamma plasma levels, which were at a maximum shortly after the end of the infusion. Isolated peripheral blood mononuclear cells obtained from patients directly after treatment with BIS-1 F(ab')2 at the 3 and 5 micrograms/kg dose level showed increased EGP-2-directed antitumor activity.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/therapy , Cell Adhesion Molecules/immunology , Immunoglobulin Fab Fragments/therapeutic use , Kidney Neoplasms/therapy , Muromonab-CD3/therapeutic use , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/adverse effects , Antibodies, Neoplasm/immunology , Carcinoma, Renal Cell/immunology , Combined Modality Therapy , Cytotoxicity, Immunologic , Dose-Response Relationship, Immunologic , Epithelial Cell Adhesion Molecule , Humans , Immunization, Passive/adverse effects , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/adverse effects , Immunoglobulin Fab Fragments/immunology , Immunologic Factors/administration & dosage , Immunologic Factors/therapeutic use , Injections, Intravenous , Injections, Subcutaneous , Interferon-gamma/blood , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Interleukin-2/therapeutic use , Kidney Neoplasms/immunology , Leukopenia/chemically induced , Muromonab-CD3/administration & dosage , Muromonab-CD3/adverse effects , Muromonab-CD3/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...