Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christoph Stein is active.

Publication


Featured researches published by Christoph Stein.


mAbs | 2012

A recombinant triplebody with specificity for CD19 and HLA-DR mediates preferential binding to antigen double-positive cells by dual-targeting

Ingo Schubert; Christian Kellner; Christoph Stein; Markus Kügler; Michael Schwenkert; Domenica Saul; Bernhard Stockmeyer; Christian Berens; Fuat S. Oduncu; Andreas Mackensen; Georg H. Fey

To test the hypothesis that dual-targeting confers the novel ability of selective binding to antigen double-positive over antigen single-positive cells, a single-chain triplebody (sctb), HLA-ds16-hu19, was produced and characterized. The molecule carries three single-chain Fv (scFv) antibody fragments in a single polypeptide chain, the two distal ones specific for the human histocompatibility protein HLA-DR and the B-lymphoid cell surface protein CD19, the central one for CD16, the human low affinity Fc-receptor FcγRIII. For comparison, the bispecific scFvs (bsscFv) hu19-ds16 and HLA-ds16 were also produced. All CD16 binding modules are disulfide-stabilized (ds). The sctb bound simultaneously to both CD19 and HLA-DR on the same cancer cell and, thus, showed functional dual-targeting. In a mixing-experiment with HLA-DR single-positive HUT-78 cells and (HLA-DR plus CD19) double-positive SEM cells, the triplebody showed preferential binding to the double-positive cells, even when the single-positive cells were present in a numerical excess of up to 20-fold. In antibody-dependent cellular cytotoxicity experiments with mononuclear cells as effector cells, the sctb promoted equal lysis of Raji cells, an antigen double-positive cell line, at 130-fold lower concentrations than the bsscFv hu19-ds16, indicating that both distal scFvs of the sctb contributed to tumor cell lysis. A panel of stably-transfected HEK293 cell lines was generated that included CD19- and HLA-DR single-positive and (HLA-DR plus CD19) double-positive lines with antigen-surface densities varying over a broad range. Using a pair of cell lines with matching densities, the sctb eliminated double-positive target cells preferentially single-positive cells. This ability of preferential or selective targeting of antigen double-positive over single-positive cells opens attractive new perspectives for the use of dual-targeting sctbs in cancer therapy.


Langmuir | 2015

Heat-Transfer-Method-Based Cell Culture Quality Assay through Cell Detection by Surface Imprinted Polymers

Kasper Eersels; Bart van Grinsven; Mehran Khorshid; Veerle Somers; Christiane Püttmann; Christoph Stein; Stefan Barth; Hanne Diliën; Gerard M. J. Bos; Wilfred T. V. Germeraad; Thomas J. Cleij; Ronald Thoelen; Ward De Ceuninck; Patrick Wagner

Previous work has indicated that surface imprinted polymers (SIPs) allow for highly specific cell detection through macromolecular cell imprints. The combination of SIPs with a heat-transfer-based read-out technique has led to the development of a selective, label-free, low-cost, and user-friendly cell detection assay. In this study, the breast cancer cell line ZR-75-1 is used to assess the potential of the platform for monitoring the quality of a cell culture in time. For this purpose, we show that the proposed methodology is able to discriminate between the original cell line (adherent growth, ZR-75-1a) and a descendant cell line (suspension growth, ZR-75-1s). Moreover, ZR-75-1a cells were cultured for a prolonged period of time and analyzed using the heat-transfer method (HTM) at regular time intervals. The results of these experiments demonstrate that the thermal resistance (Rth) signal decays after a certain number of cell culture passages. This can likely be attributed to a compromised quality of the cell culture due to cross-contamination with the ZR-75-1s cell line, a finding that was confirmed by classical STR DNA profiling. The cells do not express the same functional groups on their membrane, resulting in a weaker bond between cell and imprint, enabling cell removal by mechanical friction, provided by flushing the measuring chamber with buffer solution. These findings were further confirmed by HTM and illustrate that the biomimetic sensor platform can be used as an assay for monitoring the quality of cell cultures in time.


Langmuir | 2014

Heat-transfer resistance measurement method (HTM)-based cell detection at trace levels using a progressive enrichment approach with highly selective cell-binding surface imprints.

Karolien Bers; Kasper Eersels; Bart van Grinsven; Mat J. A. P Daemen; Jeroen F. J. Bogie; Jerome J. A. Hendriks; Evelien E. Bouwmans; Christiane Püttmann; Christoph Stein; Stefan Barth; Gerard M. J. Bos; Wilfred T. V. Germeraad; Ward De Ceuninck; Patrick Wagner

Surface-imprinted polymers allow for specific cell detection based on simultaneous recognition of the cell shape, cell size, and cell membrane functionalities by macromolecular cell imprints. In this study, the specificity of detection and the detection sensitivity for target cells within a pool of non-target cells were analyzed for a cell-specific surface-imprinted polymer combined with a heat-transfer-based read-out technique (HTM). A modified Chinese hamster ovarian cell line (CHO-ldlD) was used as a model system on which the transmembrane protein mucin-1 (MUC1) could be excessively expressed and for which the occurrence of MUC1 glycosylation could be controlled. In specific cancer cells, the overexpressed MUC1 protein typically shows an aberrant apical distribution and glycosylation. We show that surface-imprinted polymers discriminate between cell types that (1) only differ in the expression of a specific membrane protein (MUC1) or (2) only differ in the membrane protein being glycosylated or not. Moreover, surface-imprinted polymers of cells carrying different glycoforms of the same membrane protein do target both types of cells. These findings illustrate the high specificity of cell detection that can be reached by the structural imprinting of cells in polymer layers. Competitiveness between target and non-target cells was proven to negatively affect the detection sensitivity of target cells. Furthermore, we show that the detection sensitivity can be increased significantly by repetitively exposing the surface to the sample and eliminating non-specifically bound cells by flushing between consecutive cell exposures.


Cancer Letters | 2016

A novel fully-human cytolytic fusion protein based on granzyme B shows in vitro cytotoxicity and ex vivo binding to solid tumors overexpressing the epidermal growth factor receptor.

Judith Niesen; Grit Hehmann-Titt; Mira Woitok; Rolf Fendel; Stefan Barth; Rainer Fischer; Christoph Stein

Human cytolytic fusion proteins (hCFPs) offer a promising immunotherapeutic approach for the treatment of solid tumors, avoiding the immunogenicity and undesirable side-effects caused by immunotoxins derived from plants or bacteria. The well-characterized human serine protease granzyme B has already been used as a therapeutic pro-apoptotic effector domain. We therefore developed a novel recombinant hCFP (GbR201K-scFv1711) consisting of an epidermal growth factor receptor-specific human antibody fragment and a granzyme B point mutant (R201K) that is insensitive to serpin B9 (PI9), a natural inhibitor of wild-type granzyme B that is often expressed in solid tumors. We found that GbR201K-scFv1711 selectively bound to epidermoid cancer and rhabdomyosarcoma cells and was rapidly internalized by them. Nanomolar concentrations of GbR201K-scFv1711 achieved the specific killing of epidermoid cancer cells by inducing apoptosis, and similar effects were observed in rhabdomyosarcoma cells when GbR201K-scFv1711 was combined with the endosomolytic substance chloroquine. The novel hCFP was stable in serum and bound to human rhabdomyosarcoma tissue ex vivo. These data confirm that GbR201K-scFv1711 is a promising therapeutic candidate suitable for further clinical investigation.


Monoclonal antibodies in immunodiagnosis and immunotherapy | 2014

Cloning murine antibody V-genes with non-degenerate primers and conversion to a recombinant antibody format.

Magdalena Bialon; Ludmila Schellenberg; Nicolas Herzog; Stefan Kraus; Hannah Jörißen; Rainer Fischer; Christoph Stein; Jörg Nähring; Stefan Barth; Christiane Püttmann

Monoclonal antibodies are produced in cultured hybridoma cell lines, but these cells tend to be unstable; it is therefore necessary to rescue the corresponding genetic information. Here we describe an improved method for the amplification of antibody variable gene (V-gene) information from murine hybridoma cells using a panel of specific, non-degenerate primers. This primer set allows sequences to be rescued from all murine V-genes, except the lambda light chain genes, which rarely contribute to murine immune diversity. We tested the primers against a range of antibodies and recovered specific amplification products in all cases. The heavy and light chain variable regions were subsequently joined by a two-step cloning strategy or by splice overlap extension PCR.


Cancer Letters | 2016

The efficient elimination of solid tumor cells by EGFR-specific and HER2-specific scFv-SNAP fusion proteins conjugated to benzylguanine-modified auristatin F.

Mira Woitok; Diana Klose; Judith Niesen; Wolfgang Richter; Muhammad Abbas; Christoph Stein; Rolf Fendel; Magdalena Bialon; Christiane Püttmann; Rainer Fischer; Stefan Barth; Katharina Kolberg

Antibody-drug conjugates (ADCs) combine the potency of cytotoxic drugs with the specificity of monoclonal antibodies (mAbs). Most ADCs are currently generated by the nonspecific conjugation of drug-linker reagents to certain amino acid residues in mAbs, resulting in a heterogeneous product. To overcome this limitation and prepare ADCs with a defined stoichiometry, we use SNAP-tag technology as an alternative conjugation strategy. This allows the site-specific conjugation of O(6)-benzylguanine (BG)-modified small molecules to SNAP-tag fusion proteins. To demonstrate the suitability of this system for the preparation of novel recombinant ADCs, here we conjugated SNAP-tagged single chain antibody fragments (scFvs) to a BG-modified version of auristatin F (AURIF). We used two scFv-SNAP fusion proteins targeting members of the epidermal growth factor receptor (EGFR) family that are frequently overexpressed in breast cancer. The conjugation of BG-AURIF to EGFR-specific 425(scFv)-SNAP and HER2-specific αHER2(scFv)-SNAP resulted in two potent recombinant ADCs that specifically killed breast cancer cell lines by inducing apoptosis when applied at nanomolar concentrations. These data confirm that SNAP-tag technology is a promising tool for the generation of novel recombinant ADCs.


Bioconjugate Chemistry | 2016

SNAP-Tag Technology: A Useful Tool To Determine Affinity Constants and Other Functional Parameters of Novel Antibody Fragments

Judith Niesen; Markus Sack; Melanie Seidel; Rolf Fendel; Stefan Barth; Rainer Fischer; Christoph Stein

Antibody derivatives, such as the single chain fragment variable (scFv), can be developed as diagnostic and therapeutic tools in cancer research, especially in the form of fusion proteins. Such derivatives are easier to produce and modify than monoclonal antibodies (mAbs) and achieve better tissue/tumor penetration. The genetic modification of scFvs is also much more straightforward than the challenging chemical modification of mAbs. Therefore, we constructed two scFvs derived from the approved monoclonal antibodies cetuximab (scFv2112) and panitumumab (scFv1711), both of which are specific for the epidermal growth factor receptor (EGFR), a well-characterized solid tumor antigen. Both scFvs were genetically fused to the SNAP-tag, an engineered version of the human DNA repair enzyme O(6)-alkylguanine DNA alkyltransferase that allows the covalent coupling of benzylguanine (BG)-modified substrates such as fluorescent dyes. The SNAP-tag achieves controllable and irreversible protein modification and is an important tool for experimental studies in vitro and in vivo. The affinity constant of a scFv is a key functional parameter, especially in the context of a fusion protein. Therefore, we developed a method to define the affinity constants of scFv-SNAP fusion proteins by surface plasmon resonance (SPR) spectroscopy. We could confirm that both scFvs retained their functionality after fusion to the SNAP-tag in a variety of procedures and assays, including ELISA, flow cytometry, and confocal microscopy. The experimental procedures described herein, and the new protocol for affinity determination by SPR spectroscopy, are suitable for the preclinical evaluation of diverse antibody formats and derivatives.


British Journal of Haematology | 2017

Elimination of different leukaemia subtypes using novel CD89‐specific human cytolytic fusion proteins

Gerrit Gresch; Lea Schenke; Radoslav Mladenov; Stefan Zwirner; Christian Cremer; Judith Niesen; Elena Grieger; Tim H. Brümmendorf; Edgar Jost; Rainer Fischer; Bernhard Stockmeyer; Stefan Barth; Thomas Nachreiner; Christoph Stein

Colaizzo, D., Amitrano, L., Guardascione, M.A., Tiscia, G.L., D’Andrea, G., Longo, V.A., Grandone, E. & Margaglione, M. (2013) Outcome of patients with splanchnic venous thrombosis presenting without overt MPN: a role for the JAK2 V617F mutation re-evaluation. Thrombosis Research, 132, e99–e104. De Stefano, V., Vannucchi, A.M., Ruggeri, M., Cervantes, F., Alvarez-Larran, A., Iurlo, A., Randi, M.L., Pieri, L., Rossi, E., Guglielmelli, P., Betti, S., Elli, E., Finazzi, M.C., Finazzi, G., Zetterberg, E., Vianelli, N., Gaidano, G., Nichele, I., Cattaneo, D., Palova, M., Ellis, M.H., Cacciola, E., Tieghi, A., Hernandez-Boluda, J.C., Pungolino, E., Specchia, G., Rapezzi, D., Forcina, A., Musolino, C., Carobbio, A., Griesshammer, M. & Barbui, T. (2016) Splanchnic vein thrombosis in myeloproliferative neoplasms: risk factors for recurrences in a cohort of 181 patients. Blood Cancer Journal, 6, e493. Dentali, F., Squizzato, A., Brivio, L., Appio, L., Campiotti, L., Crowther, M., Grandi, A.M. & Ageno, W. (2009) JAK2V617F mutation for the early diagnosis of Phmyeloproliferative neoplasms in patients with venous thromboembolism: a meta-analysis. Blood, 113, 5617–5623. How, J., Zhou, A. & Oh, S.T. (2017) Splanchnic vein thrombosis in myeloproliferative neoplasms: pathophysiology and molecular mechanisms of disease. Therapeutic Advances in Hematology, 8, 107–118. Lamy, T., Devillers, A., Bernard, M., Moisan, A., Grulois, I., Drenou, B., Amiot, L., Fauchet, R. & Le Prise, P.Y. (1997) Inapparent polycythemia vera: an unrecognized diagnosis. American Journal of Medicine, 102, 14–20. Smalberg, J.H., Arends, L.R., Valla, D.C., Kiladjian, J.J., Janssen, H.L. & Leebeek, F.W. (2012) Myeloproliferative neoplasms in Budd-Chiari syndrome and portal vein thrombosis: a metaanalysis. Blood, 120, 4921–4928. Stein, B.L., Williams, D.M., Wang, N.Y., Rogers, O., Isaacs, M.A., Pemmaraju, N., Spivak, J.L. & Moliterno, A.R. (2010) Sex differences in the JAK2 V617F allele burden in chronic myeloproliferative disorders. Haematologica, 95, 1090–1097. Stein, B.L., Saraf, S., Sobol, U., Halpern, A., Shammo, J., Rondelli, D., Michaelis, L., Odenike, O., Rademaker, A., Zakarija, A., McMahon, B., Spivak, J.L. & Moliterno, A.R. (2013) Age-related differences in disease characteristics and clinical outcomes in polycythemia vera. Leukaemia & Lymphoma, 54, 1989–1995. Vannucchi, A.M., Antonioli, E., Guglielmelli, P., Rambaldi, A., Barosi, G., Marchioli, R., Marfisi, R.M., Finazzi, G., Guerini, V., Fabris, F., Randi, M.L., De Stefano, V., Caberlon, S., Tafuri, A., Ruggeri, M., Specchia, G., Liso, V., Rossi, E., Pogliani, E., Gugliotta, L., Bosi, A. & Barbui, T. (2007) Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood, 110, 840–846.


Oncotarget | 2016

CD64-directed microtubule associated protein tau kills leukemic blasts ex vivo

Radoslav Mladenov; Dmitrij Hristodorov; Christian Cremer; Gerrit Gresch; Elena Grieger; Lea Schenke; Diana Klose; Manal Amoury; Mira Woitok; Edgar Jost; Tim H. Brümmendorf; Rolf Fendel; Rainer Fischer; Christoph Stein; Theo Thepen; Stefan Barth

Fc gamma receptor I (FcγRI, CD64) is a well-known target antigen for passive immunotherapy against acute myeloid leukemia and chronic myelomonocytic leukemia. We recently reported the preclinical immunotherapeutic potential of microtubule associated protein tau (MAP) against a variety of cancer types including breast carcinoma and Hodgkins lymphoma. Here we demonstrate that the CD64-directed human cytolytic fusion protein H22(scFv)-MAP kills ex vivo 15–50% of CD64+ leukemic blasts derived from seven myeloid leukemia patients. Furthermore, in contrast to the nonspecific cytostatic agent paclitaxel, H22(scFv)-MAP showed no cytotoxicity towards healthy CD64+ PBMC-derived cells and macrophages. The targeted delivery of this microtubule stabilizing agent therefore offers a promising new strategy for specific treatment of CD64+ leukemia.


OncoTargets and Therapy | 2017

Comparison of a mouse and a novel human scFv-SNAP-auristatin F drug conjugate with potent activity against EGFR-overexpressing human solid tumor cells

Mira Woitok; Diana Klose; Stefano Di Fiore; Wolfgang Richter; Christoph Stein; Gerrit Gresch; Elena Grieger; Stefan Barth; Rainer Fischer; Katharina Kolberg; Judith Niesen

Antibody–drug conjugates (ADCs) can deliver toxins to specific targets such as tumor cells. They have shown promise in preclinical/clinical development but feature stoichiometrically undefined chemical linkages, and those based on full-size antibodies achieve only limited tumor penetration. SNAP-tag technology can overcome these challenges by conjugating benzylguanine-modified toxins to single-chain fragment variables (scFvs) with 1:1 stoichiometry while preserving antigen binding. Two (human and mouse) scFv-SNAP fusion proteins recognizing the epidermal growth factor receptor (EGFR) were expressed in HEK 293T cells. The purified fusion proteins were conjugated to auristatin F (AURIF). Binding activity was confirmed by flow cytometry/immunohistochemistry, and cytotoxic activity was confirmed by cell viability/apoptosis and cell cycle arrest assays, and a novel microtubule dynamics disassembly assay was performed. Both ADCs bound specifically to their target cells in vitro and ex vivo, indicating that the binding activity of the scFv-SNAP fusions was unaffected by conjugation to AURIF. Cytotoxic assays confirmed that the ADCs induced apoptosis and cell cycle arrest at nanomolar concentrations and microtubule disassembly. The SNAP-tag technology provides a platform for the development of novel ADCs with defined conjugation sites and stoichiometry. We achieved the stable and efficient linkage of AURIF to human or murine scFvs using the SNAP-tag technology, offering a strategy to improve the development of personalized medicines.

Collaboration


Dive into the Christoph Stein's collaboration.

Top Co-Authors

Avatar

Stefan Barth

University of Cape Town

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bernhard Stockmeyer

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge