Network


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

Hotspot


Dive into the research topics where Stefan Scheiblich is active.

Publication


Featured researches published by Stefan Scheiblich.


Cancer Cell | 2014

Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy

Carola Ries; Michael Cannarile; Sabine Hoves; Jörg Benz; Katharina Wartha; Valeria Runza; Flora Rey-Giraud; Leon P. Pradel; Friedrich Feuerhake; Irina Klaman; Tobin Jones; Ute Jucknischke; Stefan Scheiblich; Klaus Kaluza; Ingo H. Gorr; Antje Walz; Keelara Abiraj; Philippe Cassier; Antonio Sica; Carlos Gomez-Roca; Karin E. de Visser; Antoine Italiano; Christophe Le Tourneau; Jean-Pierre Delord; Hyam I. Levitsky; Jean-Yves Blay; Dominik Rüttinger

Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80(+) tumor-associated macrophages accompanied by an increase of the CD8(+)/CD4(+) T cell ratio. Administration of RG7155 to patients led to striking reductions of CSF-1R(+)CD163(+) macrophages in tumor tissues, which translated into clinical objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.


Protein Engineering Design & Selection | 2012

Molecular characterization of novel trispecific ErbB-cMet-IGF1R antibodies and their antigen-binding properties

Raffaella Castoldi; Ute Jucknischke; L.P. Pradel; E. Arnold; Christian Klein; Stefan Scheiblich; Gerhard Niederfellner; Claudio Sustmann

Therapeutic antibodies are well established drugs in diverse medical indications. Their success invigorates research on multi-specific antibodies in order to enhance drug efficacy by co-targeting of receptors and addressing key questions of emerging resistance mechanisms. Despite challenges in production, multi-specific antibodies are potentially more potent biologics for cancer therapy. However, so far only bispecific antibody formats have entered clinical phase testing. For future design of antibodies allowing even more targeting specificities, an understanding of the antigen-binding properties of such molecules is crucial. To this end, we have generated different IgG-like TriMAbs (trispecific, trivalent and tetravalent antibodies) directed against prominent cell surface antigens often deregulated in tumor biology. A combination of surface plasmon resonance and isothermal titration calorimetry techniques enables quantitative assessment of the antigen-binding properties of TriMAbs. We demonstrate that the kinetic profiles for the individual antigens are similar to the parental antibodies and all antigens can be bound simultaneously even in the presence of FcγRIIIa. Furthermore, cooperative binding of TriMAbs to their antigens was demonstrated. All antibodies are fully functional and inhibit receptor phosphorylation and cellular growth. TriMAbs are therefore ideal candidates for future applications in various therapeutic areas.


Molecular Cancer Therapeutics | 2013

Quantitative Chemical Proteomics Profiling Differentiates Erlotinib from Gefitinib in EGFR Wild-Type Non–Small Cell Lung Carcinoma Cell Lines

Angélique Augustin; Jens Lamerz; Hélène Meistermann; Sabrina Golling; Stefan Scheiblich; Johannes C. Hermann; Guillemette Duchateau-Nguyen; Manuel Tzouros; David W. Avila; Hanno Langen; Laurent Essioux; Barbara Klughammer

Although both erlotinib and gefitinib target the EGF receptor (EGFR), erlotinib is effective in patients with EGFR wild-type or mutated tumors, whereas gefitinib is only beneficial for patients with activating mutations. To determine whether these differences in clinical outcomes can be attributed to their respective protein interaction profiles, a label-free, quantitative chemical proteomics study was conducted. Using this method, 24 proteins were highlighted in the binding profiles of erlotinib and gefitinib. Unlike gefinitib, erlotinib displaced the ternary complex formed by integrin-linked kinase (ILK), α-parvin, and PINCH (IPP). The docking of erlotinib in the three-dimensional structure of ILK showed that erlotinib has the ability to bind to the ATP-binding site, whereas gefitinib is unlikely to bind with high affinity. As the IPP complex has been shown to be involved in epithelial-to-mesenchymal transition (EMT) and erlotinib sensitivity has been correlated with EMT status, we used a cellular model of inducible transition and observed that erlotinib prevented EMT in a more efficient way than gefitinib by acting on E-cadherin expression as well as on IPP levels. A retrospective analysis of the MERIT trial indicated that, besides a high level of E-cadherin, a low level of ILK could be linked to clinical benefit with erlotinib. In conclusion, we propose that, in an EGFR wild-type context, erlotinib may have a complementary mode of action by inhibiting IPP complex activities, resulting in the slowing down of the metastatic process of epithelial tumors. Mol Cancer Ther; 12(4); 520–9. ©2013 AACR.


Journal of Biological Chemistry | 2016

Rapid Activation of Bone Morphogenic Protein 9 by Receptor-mediated Displacement of Pro-domains

Yvonne Kienast; Ute Jucknischke; Stefan Scheiblich; Martina Thier; Mariana de Wouters; Alexander Haas; Christian Lehmann; Verena Brand; Dirk Bernicke; Konrad Honold; Stefan Lorenz

By non-covalent association after proteolytic cleavage, the pro-domains modulate the activities of the mature growth factor domains across the transforming growth factor-β family. In the case of bone morphogenic protein 9 (BMP9), however, the pro-domains do not inhibit the bioactivity of the growth factor, and the BMP9·pro-domain complexes have equivalent biological activities as the BMP9 mature ligand dimers. By using real-time surface plasmon resonance, we could demonstrate that either binding of pro-domain-complexed BMP9 to type I receptor activin receptor-like kinase 1 (ALK1), type II receptors, co-receptor endoglin, or to mature BMP9 domain targeting antibodies leads to immediate and complete displacement of the pro-domains from the complex. Vice versa, pro-domain binding by an anti-pro-domain antibody results in release of the mature BMP9 growth factor. Based on these findings, we adjusted ELISA assays to measure the protein levels of different BMP9 variants. Although mature BMP9 and inactive precursor BMP9 protein were directly detectable by ELISA, BMP9·pro-domain complex could only be measured indirectly as dissociated fragments due to displacement of mature growth factor and pro-domains after antibody binding. Our studies provide a model in which BMP9 can be readily activated upon getting into contact with its receptors. This increases the understanding of the underlying biology of BMP9 activation and also provides guidance for ELISA development for the detection of circulating BMP9 variants.


Scientific Reports | 2017

A re-engineered immunotoxin shows promising preclinical activity in ovarian cancer

Gwendlyn Kollmorgen; Klara Palme; Annette Seidl; Stefan Scheiblich; Fabian Birzele; Sabine Wilson; Christian Clemens; Edgar Voss; Martin Kaufmann; Klaus Hirzel; Natascha Rieder; Ben-Fillippo Krippendorff; Frank Herting; Gerhard Niederfellner

RG7787 is a re-engineered mesothelin-targeted immunotoxin with reduced immunogenicity composed of a humanized anti-mesothelin Fab fragment and a B-cell epitope silenced 24 kD fragment of Pseudomonas exotoxin A. High prevalence of mesothelin-positive cases and a large unmet medical need make ovarian cancer a promising indication for the clinical development of RG7787. However, ovarian cancer patients also frequently have elevated serum levels of the cancer antigen 125 (CA-125). In principle this could pose a problem, since the binding sites for CA-125 and RG7787 on mesothelin were reported to overlap. However, we show here that RG7787 can readily displace even excess amounts of CA-125 in different cellular assays. Moreover when tested in-vitro on a panel of 12 ovarian cancer cell lines, RG7787 had high cytotoxic activity on COV644, Caov-4, and SNU-119 cells and fully inhibited growth of EFO-21, KURAMOCHI, OVSAHO, and Caov-3 cells with potency values ranging from 1 to 86 pM. Finally, we evaluated the in-vivo efficacy of RG7787 in OvCa6668, a patient-derived ovarian cancer model with high levels of CA-125 expression. RG7787 had moderate monotherapy efficacy but in combination with standard chemotherapies (cisplatin, paclitaxel) achieved pronounced tumor regressions. In summary our data support clinical testing of RG7787 in ovarian cancer.


Cancer Research | 2015

Abstract 5321: Preclinical validation for treatment with RG7787 in ovarian cancer

Gwendlyn Kollmorgen; Klara Palme; Annette Seidl; Stefan Scheiblich; Christian Clemens; Edgar Voss; Martin Kaufmann; Klaus Hirzel; Pamela Wilfert; Moritz Marcinowski; Bernd Satzinger; Frank Herting; Gerhard Niederfellner

RG7787 is composed of a Fab fragment from an anti-MSLN antibody fused to a de-immunized and truncated pseudomonas endotoxin A variant. Once RG7787 is bound and internalized by MSLN positive cells, the toxin is transported to the cytosol, where it inhibits protein synthesis, eventually causing tumor cell death. Mesothelin (MSLN) is a tumor specific differentiation antigen. On normal tissue, its expression is restricted to differentiated mesothelial cells that line as single cell layer body cavities and major organs (e.g. pleura, pericardium, and peritoneum). In cancer, MSLN is highly expressed not only on mesotheliomas but also on a number of other types of solid tumors like ovarian and pancreatic cancer. In both indications patients frequently also have significant serum levels of the cancer antigen-125 (CA-125), sometimes even >1000 U/ml. MSLN has been described to bind CA-125 and this interaction has been suggested to play a role for the ability of cancer cells to metastasize e.g. to the peritoneum. The region in MSLN that is responsible for its interaction with CA-125 has been reported to overlap with the binding epitope of RG7787 potentially resulting in competition between CA-125 and RG7787 for binding to MSLN. The high percentage of MSLN positive cases as well as a clear unmet medical need makes ovarian and pancreatic cancer promising indications for clinical development of RG7787. We investigated, whether abundance of CA-125 can negatively affect ability of RG7787 to bind and be taken up by tumor cells thereby antagonizing RG7787 treatment. We found that indeed RG7787 competes with CA-125 for binding to mesothelin. However, in SPR experiments, the interaction of MSLN with CA-125 was not strong enough to block binding of the anti-MSLN Fab moiety that targets RG7787 to tumor cells. At 20°C the absolute affinity (KD) of RG7787 for human MSLN was determined to be 12.5 pM using two orthogonal “affinity in solution” methods, ELISA and SPR. In agreement with such high affinity binding, we found that adding soluble CA-125 to cell viability assays did not reduce the cytotoxic potency of RG7787. In one set of assays, ascites fluid containing a 10 fold excess of CA-125 compared to the average levels observed in sera of ovarian patients was used for competition. In another set of assays, we used a 100 fold excess of a truncated recombinant CA-125 fragment, containing the domain that interacts with MSLN. Neither the ascites fluid, nor the recombinant CA-125 fragment had any effect on the in-vitro potency of RG7787, indicating that soluble CA-125 levels in patients will not antagonize RG7787 treatment. Finally we demonstrated in cell-cell interaction assays that RG7787 can efficiently block the attachment of MSLN positive cells to OVCAR3 cells that express high levels of CA-125 on their surface. Based on these encouraging data we are currently evaluating in-vivo efficacy of RG7787 in a patient-derived ovarian cancer model. Citation Format: Gwendlyn Kollmorgen, Klara Palme, Annette Seidl, Stefan Scheiblich, Christian Clemens, Edgar Voss, Martin Kaufmann, Klaus Hirzel, Pamela Wilfert, Moritz Marcinowski, Bernd Satzinger, Frank Herting, Gerhard Niederfellner. Preclinical validation for treatment with RG7787 in ovarian cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5321. doi:10.1158/1538-7445.AM2015-5321


Cancer Research | 2015

Abstract PR04: Targeting tumor-asoociated macrophages with a novel anti-CSF1R antibody in cancer patients

Michael Cannarile; Sabine Hoves; Ann-Marie Broeske; Joerg Benz; Katharina Wartha; Valeria Runza; Flora Rey-Giraud; Leon P. Pradel; Friedrich Feuerhake; Irina Klaman; Tobin Jones; Ute Jucknischke; Stefan Scheiblich; Ingo H. Gorr; Antje Walz; Keelara Abiraj; Philippe Cassier; Antonio Sica; Carlos Gomez-Roca; Christophe Le Tourneau; Jean-Pierre Delord; Antoine Italiano; Hyam I. Levitsky; Jean-Yves Blay; Dominik Ruettinger; Carola Ries

Myeloid cells represent the most abundant immune cell type within the tumor microenvironment of certain tumor entities, including tumor associated macrophages (TAMs). Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for TAMs is macrophage colony stimulating factor 1 (CSF1). We generated a monoclonal antibody (RG7155) that binds to the secondary dimerization interface of CSF1 receptor (CSF1R) as a specific and potent allosteric inhibitor. In vitro, RG7155 treatment results in cell death of CSF1-differentiated macrophages. In animal models, CSF1R inhibition reduced the F4/80+ TAMs infiltrate by 90% and was accompanied by an increase of the CD8+/CD4+ T cell ratio. The ability of RG7155 to reduce TAMs is currently evaluated in a first-in-man phase I clinical study in patients suffering either from pigmented villonodular synovitis (PVNS), a neoplastic disorder characterized by CSF1 overexpression, or other tumor entities. The associated biomarker program involves mandatory paired pre- and on-treatment biopsies of tumor and surrogate skin tissue as well as pharmacodynamic marker assessment in circulating blood. In patients treated with RG7155 an increase of CSF1 associated with a sustained decrease of CD14+CD16+ alternatively activated monocytes in peripheral blood was detected. In PVNS patients administration of RG7155 led to striking reductions of CSF1R+ and CD163+ macrophages in tumor tissue resulting in objective clinical responses according to RECIST (Response Evaluation Criteria in Solid Tumors) in 5 out of 6 patients. All six evaluable PVNS patients showed partial metabolic response in FDG-PET imaging and significant symptomatic improvement as early as 4 weeks after treatment initiation. Furthermore, TAM reduction was also observed in paired tumor samples of patients with various advanced solid malignancies, suggesting broad applicability of this therapeutic approach. This abstract is also presented as Poster A50. Citation Format: Michael Cannarile, Sabine Hoves, Ann-Marie Broeske, Joerg Benz, Katharina Wartha, Valeria Runza, Flora Rey-Giraud, Leon P. Pradel, Friedrich Feuerhake, Irina Klaman, Tobin Jones, Ute Jucknischke, Stefan Scheiblich, Ingo H. Gorr, Antje Walz, Keelara Abiraj, Philippe Cassier, Antonio Sica, Carlos Gomez-Roca, Christophe Le Tourneau, Jean-Pierre Delord, Antoine Italiano, Hyam Levitsky, Jean-Yves Blay, Dominik Ruettinger, Carola H. Ries. Targeting tumor-asoociated macrophages with a novel anti-CSF1R antibody in cancer patients. [abstract]. In: Abstracts: AACR Special Conference on Cellular Heterogeneity in the Tumor Microenvironment; 2014 Feb 26-Mar 1; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(1 Suppl):Abstract nr PR04. doi:10.1158/1538-7445.CHTME14-PR04


Cancer Research | 2011

Abstract LB-397: Functional characterization of IGF-1R antibodies and possible implications for clinical safety and efficacy

Klaus-Peter Kuenkele; Marco H. Hofmann; Stefan Scheiblich; Katharina Wartha; Christian Klein

Background This is the first study performing a head to head comparison of monoclonal IGF-1R antibodies (mAb) based on published sequences for therapeutic mAbs from Pfizer (CP751,871, IgG2 kappa), Amgen (AMG479, IgG1 lambda), Merck (h7C10, cloned to R1507 backbone), Imclone (IMC-A12, IgG1 lambda) and Roche (R1507, IgG1). Methods In this study, sequence information for IGF-1R mAbs was extracted from patents and used to clone and transiently express IgGs (*=re-synthesized) in HEK293F cells. In vitro assays for ligand binding, IGF-1R auto-phosphorylation, IGF-1R downregulation, IR co-downregulation, and affinity analyses (Biacore) were used. In vivo comparison was done in BxPC3 xenograft mouse model. Results All antibodies inhibit IGF-1 binding and signaling at low nanomolar levels. While IMC-A12* and R1507 also prevent IGF-2 binding to IGF-1R and subsequent receptor activation, CP751,871* and h7C10* do not inhibit IGF-2 binding and have only limited impact (55%/30% inhibition) on IGF-2 signaling. Analysis of IGF-1R phosphorylation in 0.5 % FCS medium revealed that all antibodies except R1507 exert agonistic activity. Interestingly, Fab fragments of agonistic mAbs became antagonistic, indicating that bivalent binding is necessary for agonistic effects. All mAb (200nM, 24h treatment of MCF-7 cells) with the exception of AMG479* efficiently downregulated 78–82% the IGF-1R. Analysis of the same cell lysates revealed however striking differences in IR co-downregulation, a mechanism discussed as possible cause of clinical hyperglycemia. R1507 had the least side effects on Insulin co-downregulation (9%) compared to h7C10* (15%), CP751,871* (23%) and IMC-A12* (46%). Differences were also seen in the binding kinetics. Both AMG479* and R1507 showed faster k off rates resulting in shorter retention times at the receptor. Since k on /k off rates are discussed to influence tumor penetration (1), we compared downregulation of IGF-1R by R1507 and CP751,871* in xenograft tumors. Although both mAbs downregulate IGF-1R in vitro to the same extent, R1507 was significantly more effective in the in-vivo setting. Conclusion The head to head comparison of IGF-1R mAbs revealed differences in regard to binding properties, tumor penetration, IR codownregulation, and inhibition of signaling via the ligand IGF-2. Reference List 1. Adams, G. P., Schier, R., McCall, A. M., Simmons, H. H., Horak, E. M., Alpaugh, R. K., Marks, J. D., and Weiner, L. M. (2001) Cancer Res. 61 , 4750–4755 Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-397. doi:10.1158/1538-7445.AM2011-LB-397


Journal of Molecular Biology | 2005

Crystal structures of active SRC kinase domain complexes

Christine B. Breitenlechner; Norman Kairies; Konrad Honold; Stefan Scheiblich; Hans Koll; Eva Greiter; Stefan Koch; Wolfgang Schäfer; Robert Huber; Richard A. Engh


Archive | 2007

Heteroaryl derivatives as protein kinase inhibitors

Konrad Honold; Wolfgang Schaefer; Stefan Scheiblich

Collaboration


Dive into the Stefan Scheiblich's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge