Network


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

Hotspot


Dive into the research topics where Holger Hess-Stumpp is active.

Publication


Featured researches published by Holger Hess-Stumpp.


Cancer Research | 2008

Inhibition of Multiple Vascular Endothelial Growth Factor Receptors (VEGFR) Blocks Lymph Node Metastases but Inhibition of VEGFR-2 Is Sufficient to Sensitize Tumor Cells to Platinum-Based Chemotherapeutics

Patrizia Sini; Ivana Samarzija; Fabienne Baffert; Amanda Littlewood-Evans; Christian Schnell; Andreas Theuer; Sven Christian; Anja M. Boos; Holger Hess-Stumpp; John A. Foekens; Buddy Setyono-Han; Jeanette Marjorie Wood; Nancy E. Hynes

Vascular endothelial growth factor receptors (VEGFR) have important roles in cancer, affecting blood and lymphatic vessel functionality as well as tumor cells themselves. We compared the efficacy of a VEGFR tyrosine kinase inhibitor, PTK787/ZK222584 (PTK/ZK), which targets the three VEGFRs, with blocking antibodies directed against VEGFR-2 (DC101) or VEGF-A (Pab85618) in a metastatic melanoma model. Although all inhibitors exerted comparable effects on primary tumor growth, only PTK/ZK significantly reduced lymph node metastasis formation. A comparable decrease in lymphatic vessel density following blockade of VEGFR-2 (DC101) or the three VEGFRs (PTK/ZK) was observed in the metastases. However, the functionality of lymphatics surrounding the primary tumor was more significantly disrupted by PTK/ZK, indicating the importance of multiple VEGFRs in the metastatic process. The antimetastatic properties of PTK/ZK were confirmed in a breast carcinoma model. B16/BL6 tumor cells express VEGF ligands and their receptors. Blockade of a VEGFR-1 autocrine loop with PTK/ZK inhibited tumor cell migration. Furthermore, the tumor cells also showed enhanced sensitivity to platinum-based chemotherapy in combination with PTK/ZK, indicating that autocrine VEGFRs are promoting tumor cell migration and survival. In summary, our results suggest that, in addition to blocking angiogenesis, combined inhibition of the three VEGFRs may more efficiently target other aspects of tumor pathophysiology, including lymphatic vessel functionality, tumor cell dissemination, survival pathways, and response to chemotherapeutic compounds.


Cancer Medicine | 2013

BAY 87-2243, a highly potent and selective inhibitor of hypoxia-induced gene activation has antitumor activities by inhibition of mitochondrial complex I

Peter Ellinghaus; Iring Heisler; Kerstin Unterschemmann; Michael Haerter; Hartmut Beck; Susanne Greschat; Alexander Ehrmann; Holger Summer; Ingo Flamme; Felix Oehme; Karl-Heinz Thierauch; Martin Michels; Holger Hess-Stumpp; Karl Ziegelbauer

The activation of the transcription factor hypoxia‐inducible factor‐1 (HIF‐1) plays an essential role in tumor development, tumor progression, and resistance to chemo‐ and radiotherapy. In order to identify compounds targeting the HIF pathway, a small molecule library was screened using a luciferase‐driven HIF‐1 reporter cell line under hypoxia. The high‐throughput screening led to the identification of a class of aminoalkyl‐substituted compounds that inhibited hypoxia‐induced HIF‐1 target gene expression in human lung cancer cell lines at low nanomolar concentrations. Lead structure BAY 87‐2243 was found to inhibit HIF‐1α and HIF‐2α protein accumulation under hypoxic conditions in non‐small cell lung cancer (NSCLC) cell line H460 but had no effect on HIF‐1α protein levels induced by the hypoxia mimetics desferrioxamine or cobalt chloride. BAY 87‐2243 had no effect on HIF target gene expression levels in RCC4 cells lacking Von Hippel–Lindau (VHL) activity nor did the compound affect the activity of HIF prolyl hydroxylase‐2. Antitumor activity of BAY 87‐2243, suppression of HIF‐1α protein levels, and reduction of HIF‐1 target gene expression in vivo were demonstrated in a H460 xenograft model. BAY 87‐2243 did not inhibit cell proliferation under standard conditions. However under glucose depletion, a condition favoring mitochondrial ATP generation as energy source, BAY 87‐2243 inhibited cell proliferation in the nanomolar range. Further experiments revealed that BAY 87‐2243 inhibits mitochondrial complex I activity but has no effect on complex III activity. Interference with mitochondrial function to reduce hypoxia‐induced HIF‐1 activity in tumors might be an interesting therapeutic approach to overcome chemo‐ and radiotherapy‐resistance of hypoxic tumors.


Acta Neuropathologica | 2017

Pan-mutant IDH1 inhibitor BAY 1436032 for effective treatment of IDH1 mutant astrocytoma in vivo.

Stefan Pusch; Sonja Krausert; Viktoria Fischer; Jörg Balss; Martina Ott; Daniel Schrimpf; David Capper; Felix Sahm; Jessica Eisel; Ann Christin Beck; Manfred Jugold; Viktoria Eichwald; Stefan Kaulfuss; Olaf Panknin; Hartmut Rehwinkel; Katja Zimmermann; Roman Hillig; Judith Guenther; Luisella Toschi; Roland Neuhaus; Andrea Haegebart; Holger Hess-Stumpp; Markus Bauser; Wolfgang Wick; Andreas Unterberg; Christel Herold-Mende; Michael Platten; Andreas von Deimling

Mutations in codon 132 of isocitrate dehydrogenase (IDH) 1 are frequent in diffuse glioma, acute myeloid leukemia, chondrosarcoma and intrahepatic cholangiocarcinoma. These mutations result in a neomorphic enzyme specificity which leads to a dramatic increase of intracellular d-2-hydroxyglutarate (2-HG) in tumor cells. Therefore, mutant IDH1 protein is a highly attractive target for inhibitory drugs. Here, we describe the development and properties of BAY 1436032, a pan-inhibitor of IDH1 protein with different codon 132 mutations. BAY 1436032 strongly reduces 2-HG levels in cells carrying IDH1-R132H, -R132C, -R132G, -R132S and -R132L mutations. Cells not carrying IDH mutations were unaffected. BAY 1436032 did not exhibit toxicity in vitro or in vivo. The pharmacokinetic properties of BAY 1436032 allow for oral administration. In two independent experiments, BAY 1436032 has been shown to significantly prolong survival of mice intracerebrally transplanted with human astrocytoma carrying the IDH1R132H mutation. In conclusion, we developed a pan-inhibitor targeting tumors with different IDH1R132 mutations.


ChemMedChem | 2014

Inhibition of hypoxia-induced gene transcription by substituted pyrazolyl oxadiazoles: initial lead generation and structure-activity relationships.

Michael Härter; Karl-Heinz Thierauch; Stephen Boyer; Ajay Bhargava; Peter Ellinghaus; Hartmut Beck; Susanne Greschat-Schade; Holger Hess-Stumpp; Kerstin Unterschemmann

The transcription factors hypoxia‐inducible factor‐1 and ‐2 (HIF‐1 and HIF‐2) orchestrate a multitude of processes that allow tumor cells to survive under conditions of low oxygen and nutrients, and that lead to resistance to some apoptotic pathways and facilitate invasion and metastasis. Therefore, inhibition of transactivation by HIF has become an attractive target in cancer research. Herein we present the results of a cell‐based screening approach that led to the discovery of substituted 1H‐pyrazole‐3‐carboxamides. Chemical optimization of the hit class with respect to potency and metabolic stability is described; it resulted in novel 5‐(1H‐pyrazol‐3‐yl)‐1,2,4‐oxadiazoles that inhibit the hypoxia‐induced accumulation of HIF‐1α and HIF‐2α. The HIF inhibitory potency in the screening cell system was improved from IC50 190 to 0.7 nM, and significant parts of the SAR are disclosed. For a key compound, the ability to suppress the hypoxia‐induced expression of HIF target genes was studied in A549 human lung adenocarcinoma cells. The same compound shows a favorable pharmacokinetic profile in rats after i.v. and p.o. administration.


Cancer Research | 2014

Abstract 1739: Preclinical profile of BAY 1163877 - a selective pan-FGFR inhibitor in phase 1 clinical trial

Melanie Heroult; Peter Ellinghaus; Christian Sieg; Dirk Brohm; Sylvia Gruenewald; Marie-Pierre Collin; Ulf Boemer; Mario Lobell; Walter Huebsch; Matthias Ocker; Stuart Ince; Andrea Haegebarth; Rolf Jautelat; Holger Hess-Stumpp; Michael Brands; Karl Ziegelbauer

Fibroblast growth factors (FGFs) orchestrate a variety of cellular functions by binding to their transmembrane tyrosine-kinase receptors (FGFR1-4) and activating downstream signaling pathways. FGF signaling has been demonstrated to be altered in a high proportion of cancers, with activating mutations and/or overexpression of FGFRs frequently observed in lung, gastric, breast and urothelial tumors. Therefore, targeting FGFRs using selective FGFR inhibitors is an attractive therapeutic approach to treat cancer patients. BAY 1163877 is as an orally available, selective and potent inhibitor of FGFR-1, -2 and -3 kinase activity. BAY 1163877 has been advanced through preclinical development and we disclose here the first details of its preclinical profile. BAY 1163877 inhibited FGFR-1, -2, -3 kinase activity in the nanomolar range and demonstrated a kinase selectivity profile for FGFR-1, -2 and -3 over 222 kinases tested. BAY 1163877 inhibited proliferation of various cancer cell lines in vitro and phosphorylation of downstream signaling molecules. BAY 1163877 was also tested in vivo in monotherapy and combination therapy on various human xenografts and syngeneic tumors and inhibited growth of tumors presenting at least one FGFR alteration.Overall, the in vitro and in vivo studies confirm that the FGFR inhibitor BAY 1163877 is a potent and selective inhibitor of altered FGFRs pathways in cancer models. A Phase 1 clinical trial (NCT01976741) has been initiated. Citation Format: Melanie Heroult, Peter Ellinghaus, Christian Sieg, Dirk Brohm, Sylvia Gruenewald, Marie-Pierre Collin, Ulf Boemer, Mario Lobell, Walter Huebsch, Matthias Ocker, Stuart Ince, Andrea Haegebarth, Rolf Jautelat, Holger Hess-Stumpp, Michael Brands, Karl Ziegelbauer. Preclinical profile of BAY 1163877 - a selective pan-FGFR inhibitor in phase 1 clinical trial. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1739. doi:10.1158/1538-7445.AM2014-1739


Cancer Research | 2016

Abstract 2645: BAY 1436032: A highly selective, potent and orally available inhibitor of mutant forms of IDH1

Olaf Panknin; Stefan Pusch; Lena Herbst; Stefan Kaulfuss; Katja Zimmermann; Hartmut Rehwinkel; Roland Neuhaus; Sven Ring; Michael Brüning; Claudia Stark; Katja Prelle; Martin Michels; Michael Jeffers; Holger Hess-Stumpp; Karl Ziegelbauer; Michael Brands; Alwin Krämer; Andreas von Deimling

Isocitrate dehydrogenase 1 (IDH1) is a metabolic enzyme that is frequently mutated in certain cancers, with incidence rates ranging from 7-90% for glioma, chondrosarcoma, intrahepatic cholangiocarcinoma and AML. Wildtype IDH1 (wtIDH1) catalyzes the conversion of isocitrate to α-ketoglutarate (αKG), while tumor-associated mutant IDH1 (mIDH1) catalyzes a rogue reaction: the production of 2-hydroxyglutarate (2-HG) from αKG. 2-HG therefore represents an “oncometabolite” that is believed to play a role in cancer by interfering with αKG-dependent enzymes, which in turn causes hypermethylation of histones/DNA and a block of normal cellular differentiation. Mutant IDH1 is a “driver” oncogene and the inhibition of this altered enzyme will decrease the growth of mIDH1 dependent tumors. We report for the first time the preclinical profile and structure of BAY 1436032, a novel selective mIDH1 inhibitor. An optimization program based on a high throughput screening resulted in the identification of the clinical candidate BAY 1436032 for the treatment of mIDH1 dependent cancer. BAY 1436032 is a double-digit nanomolar and selective pan-inhibitor of the enzymatic activity of various IDH1-R132X mutants in vitro and displayed potent inhibition of 2-HG release (nanomolar range) in patient derived and engineered cell lines expressing different IDH1 mutants. In line with the proposed mode of action, a concentration-dependent lowering of 2-HG was observed in vitro accompanied by differentiation and maturation of mIDH1 tumor cells. Furthermore, BAY 1436032 showed a favourable selectivity profile against wtIDH1/2 and a large panel of off-targets in vitro. To the best of our knowledge we were able to show for the first time single agent in vivo efficacy in mIDH1 patient derived glioma and intrahepatic cholangiocarcinoma solid tumor models with this clinical candidate along with monitoring of intratumoral 2-HG levels as a predictive biomarker. The BBB penetration profile of BAY 1436032 is further supported by preclinical data on in vivo brain-plasma ratios. In conclusion, our data provide in vitro and in vivo proof of concept for BAY 1436032 as a potent and highly selective inhibitor of mutant forms of IDH1. The start of a Phase I study with BAY 1436032 is currently in preparation to determine the safety, tolerability, pharmacokinetics and preliminary anti-tumor and pharmacodynamic biomarker responses in patients with solid tumors. Citation Format: Olaf Panknin, Stefan Pusch, Lena Herbst, Stefan Kaulfuss, Katja Zimmermann, Hartmut Rehwinkel, Roland Neuhaus, Sven Ring, Michael Bruning, Claudia Stark, Katja Prelle, Martin Michels, Michael Jeffers, Holger Hess-Stumpp, Karl Ziegelbauer, Michael Brands, Alwin Kramer, Andreas von Deimling. BAY 1436032: A highly selective, potent and orally available inhibitor of mutant forms of IDH1. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2645.


Cancer Research | 2014

Abstract 1026: Novel Tie2 inhibitor with in vivo efficacy in disseminated hematological tumor models in mice

Sylvia Gruenewald; Julia Schueler; Michael Haerter; Frank Suessmeier; Kerstin Klingner; Ulf Boemer; Stefan Kaulfuss; Alexander Walter; Mario Lobell; Ingo Hartung; Bernd Buchmann; Dieter Heldmann; Holger Hess-Stumpp; Karl Ziegelbauer

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The receptor tyrosine kinase Tie2 is predominantly expressed in the endothelium but has also been identified on primitive hematopoietic stem cells, monocyte and macrophage subclasses, as well as on glioma or hematological tumor cells. Based on its expression in many patient-derived leukemic blasts inhibition of the Tie2 pathway may provide an attractive opportunity for therapeutic intervention in leukemias. In this study we report the pharmacological profile of a novel, highly potent and orally available Tie2 inhibitor (BAY-Tie2). The discovery and design process leading to BAY-Tie2 was performed with the goal of sparing other angiogenic RTKs, such as VEGFRs, FGFRs or PDGFRs. BAY-Tie2 is based on a novel imidazopyrazole core, combined with a SF5-substituted phenyl ring that fills the deep DFG-out pocket. BAY-Tie2 binds to Tie2 with a Kd value of 1.6 nM and is selective against VEGFR2 (Kd of 1600 nM), FGFR1 (<30% inhibition at 1 µM), FGFR2/3/4 (<10% inhibition at 1 µM) and PDGFRα/β (<30% inhibition at 100 nM). BAY-Tie2 potently inhibits Tie2 autophosphorylation in recombinant CHO-Tie2 and primary human umbilical vein endothelial cells (HUVEC) with IC50 values of 0.7 and 1.3 nM. Consistently, BAY-Tie2 was shown to inhibit Tie2 phosphorylation in vivo by analyzing angiopoietin-1 induced Tie2 phosphorylation status in extracts of murine lungs from BAY-Tie2-treated mice. In subcutaneous xenograft models of highly angiogenic tumors, BAY-Tie2 reduced tumor growth and showed evidence for potential combination benefit with anti-VEGF therapy. In order to explore the potential of a Tie2 inhibitor beyond affecting angiogenesis, we established disseminated leukemia models, using Tie2-expressing cell lines, such as the CML cell lines MEG-01 and EM-2. Both cell lines engrafted predominantly in bone marrow and spleen. Treatment started 3 days after i.v. cell implantation with either BAY-Tie2 or cytarabine and was well tolerated. Efficacy was monitored by a) inhibition of disease progression, b) weekly fluorescence-based in vivo imaging (IVI) using an Alexa750-labeled anti-human CD33 antibody, and c) q-RT-PCR specific for BCR-ABL and hCD45 in murine peripheral blood. BAY-Tie2 inhibited disease progression comparable to cytarabine. Tumor load measured by IVI was reduced in BAY-Tie2 treated groups by 45% in the MEG-01 and by 65% in the EM-2 model compared with the untreated control, very similar to the cytotoxic treatment with cytarabine. Quantitative RT-PCR on peripheral blood revealed that BAY-Tie2 and cytarabine delayed the appearance of circulating tumor cells in both CML models. These data demonstrate that BAY-Tie2 is an orally active Tie2 inhibitor that may have therapeutic benefit not only in angiogenic tumors but also in hematological, Tie2-expressing malignancies. Citation Format: Sylvia Gruenewald, Julia Schueler, Michael Haerter, Frank Suessmeier, Kerstin Klingner, Ulf Boemer, Stefan Kaulfuss, Alexander Walter, Mario Lobell, Ingo V. Hartung, Bernd Buchmann, Dieter Heldmann, Holger Hess-Stumpp, Karl Ziegelbauer. Novel Tie2 inhibitor with in vivo efficacy in disseminated hematological tumor models in mice. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1026. doi:10.1158/1538-7445.AM2014-1026


Cancer Research | 2013

Abstract 4953: Stromal endosialin modulates the proinflammatory tumor microenvironment and is crucial for the growth of orthotopic pancreatic tumors.

Sven Christian; Vijaysahankar Sivanandam; Michael Strerath; Heiner Apeler; Holger Hess-Stumpp; Hellmut G. Augustin

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Endosialin (CD248), a transmembrane protein expressed in stromal fibroblasts and activated pericytes during embryonic development, is expressed in human carcinomas. Although its function is unknown, due to its highly specific expression pattern endosialin represents a promising anti-tumor target. To unravel the role of endosialin during tumor progression and metastasis, we employed systematic loss-of-function approaches in stromal fibroblasts in vitro as well as in subcutaneous and orthotopic tumors in vivo. These experiments revealed an important role of endosialin in regulating fibroblast proliferation and in orchestrating an inflammatory response by modulating the secretion of inflammatory cytokines (IL6) in stromal fibroblasts. The proinflammatory environment created by activated fibroblasts is thought to support tumor growth and metastasis in vivo. Experiments in endosialin-deficient mice confirmed an important role of endosialin in tumor progression in highly stroma-recruiting orthotopic pancreatic tumor models. Finally, lung metastases were dramatically decreased in endosialin-deficient mice supporting a role of a proinflammatory tumor stroma in tumor dissemination. Taken together, our experiments identify endosialin as an important modulator of stromal responses that are crucial for tumor progression and metastasis. Endosialin may therefore represent a novel target for development of anticancer therapies for solid tumors. Citation Format: Sven Christian, Vijaysahankar Sivanandam, Michael Strerath, Heiner Apeler, Holger Hess-Stumpp, Hellmut G. Augustin. Stromal endosialin modulates the proinflammatory tumor microenvironment and is crucial for the growth of orthotopic pancreatic tumors. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4953. doi:10.1158/1538-7445.AM2013-4953


Cancer Research | 2012

Abstract LB-244: BAY 87-2243, an inhibitor of HIF-1α - induced gene activation, showed promising anti-tumor efficacy in combination with anti-angiogenic therapy and irradiation in preclinical tumor models

Holger Hess-Stumpp

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Hypoxia is a hallmark of solid tumors and is associated with local invasion, metastatic spread and resistance to radio- as well as chemotherapy (Vaupel et al., 2004). Furthermore, hypoxia constitutes an independent negative prognostic factor in a diverse range of malignant tumors (Harris, 2002; Vaupel et al., 2004). The cellular adaptation to hypoxia is mediated by a heterodimeric transcription factor hypoxia inducible factor-1 (HIF1). HIF-1α, one subunit of HIF, is constitutively degraded under normoxic conditions, but is stabilized and activated in hypoxic regions of tumors. Nuclear activated HIF-1 controls the expression of >100 genes involved in cellular energy metabolism, neoangiogenesis, anti-apoptotic and pro-proliferative mechanisms promoting invasion and metastasis. Recently, we showed that BAY 87-2243, a small molecule inhibitor of hypoxia-induced HIF-1 activation that specifically suppressed hypoxia-induced HIF-1α stabilization is able to achieve very promising results in combination with standard treatments in several preclinical tumor xenograft models. Here, we present outstanding preclinical anti-tumor efficacy of BAY 87-2243 in combination with anti-angiogenic therapy and irradiation in xenografts of various histological type. BAY 87-2243 was dosed continuously in combination with the tyrosine kinase inhibitors regorafenib as well as sorafenib and the anti-VEGF antibody, bevacizumab. The combinations produced long-term tumor growth control or tumor stasis of either cell line derived or patient-derived subcutaneous colorectal carcinoma xenografts (HCT116, HT29, LoVo). In a fully established lung carcinoma model (A549) the combination treatment of BAY 87-2243 with regorafenib resulted in slight regression of tumor growth that was maintained for up to 18 days. In a prostate carcinoma model (PC-3) a combination of BAY 87-2243 and regorafenib resulted in an additive improvement of the tumor growth inhibition as compared to either agents alone. A first combination treatment of BAY 87-2243 and a single subcurative irradiation dose (8 Gy) in a radioresistant, severely hypoxic head and neck squamous cell carcinoma xenograft (UT-SCC-5) led to tumor regression, which did not occur in the respective single-agent/radiation treatment. These promising preclinical results suggest that inhibition of HIF-1 by BAY 87-2243 in particular in combination with anti-angiogenic therapy and irradiation is an innovative approach to cancer therapy with the potential to overcome hypoxia/HIF-induced tumor resistance mechanisms. Harris AL (2002) Hypoxia-a key regulatory factor in tumor growth. Nat Rev Cancer 2: 38-47 Vaupel P, Mayer A, Hockel M (2004) Tumor hypoxia and malignant progression. Methods Enzymol 381:335-354 Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-244. doi:1538-7445.AM2012-LB-244


Archive | 2003

VEGFR-2 and VEGFR-3 inhibitory anthranilamide pyridines

Andreas Huth; Ludwig Zorn; Martin Krüger; Stuart Ince; Karl-Heinz Thierauch; Andreas Menrad; Martin Haberey; Holger Hess-Stumpp

Collaboration


Dive into the Holger Hess-Stumpp's collaboration.

Top Co-Authors

Avatar

Dirk Kosemund

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lars Wortmann

Bayer Schering Pharma AG

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Volker Schulze

Bayer Schering Pharma AG

View shared research outputs
Top Co-Authors

Avatar

Imadul Islam

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Karl Ziegelbauer

Bayer HealthCare Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge