Network


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

Hotspot


Dive into the research topics where Marcus Bauser is active.

Publication


Featured researches published by Marcus Bauser.


ChemMedChem | 2016

Identification and Optimization of the First Highly Selective GLUT1 Inhibitor BAY‐876

Holger Siebeneicher; Arwed Cleve; Hartmut Rehwinkel; Roland Neuhaus; Iring Heisler; Thomas Müller; Marcus Bauser; Bernd Buchmann

Despite the long‐known fact that the facilitative glucose transporter GLUT1 is one of the key players safeguarding the increase in glucose consumption of many tumor entities even under conditions of normal oxygen supply (known as the Warburg effect), only few endeavors have been undertaken to find a GLUT1‐selective small‐molecule inhibitor. Because other transporters of the GLUT1 family are involved in crucial processes, these transporters should not be addressed by such an inhibitor. A high‐throughput screen against a library of ∼3u2005million compounds was performed to find a small molecule with this challenging potency and selectivity profile. The N‐(1H‐pyrazol‐4‐yl)quinoline‐4‐carboxamides were identified as an excellent starting point for further compound optimization. After extensive structure–activity relationship explorations, single‐digit nanomolar inhibitors with a selectivity factor of >100 against GLUT2, GLUT3, and GLUT4 were obtained. The most promising compound, BAY‐876 [N4‐[1‐(4‐cyanobenzyl)‐5‐methyl‐3‐(trifluoromethyl)‐1H‐pyrazol‐4‐yl]‐7‐fluoroquinoline‐2,4‐dicarboxamide], showed good metabolic stability inu2005vitro and high oral bioavailability inu2005vivo.


eLife | 2018

Science Forum: Donated chemical probes for open science

Susanne Müller; Suzanne Ackloo; C.H. Arrowsmith; Marcus Bauser; Jeremy L Baryza; Julian Blagg; Jark Böttcher; C. Bountra; Peter J. Brown; Mark E Bunnage; Adrian Carter; David R Damerell; Volker Dötsch; David H. Drewry; A. Edwards; James P. Edwards; Jon M Elkins; Christian Fischer; Stephen V. Frye; Andreas Gollner; Charles E Grimshaw; Adriaan P. Ijzerman; Thomas Hanke; Ingo Hartung; Steve Hitchcock; Trevor Howe; Terry V Hughes; Stefan Laufer; Volkhart Mj Li; Spiros Liras

Potent, selective and broadly characterized small molecule modulators of protein function (chemical probes) are powerful research reagents. The pharmaceutical industry has generated many high-quality chemical probes and several of these have been made available to academia. However, probe-associated data and control compounds, such as inactive structurally related molecules and their associated data, are generally not accessible. The lack of data and guidance makes it difficult for researchers to decide which chemical tools to choose. Several pharmaceutical companies (AbbVie, Bayer, Boehringer Ingelheim, Janssen, MSD, Pfizer, and Takeda) have therefore entered into a pre-competitive collaboration to make available a large number of innovative high-quality probes, including all probe-associated data, control compounds and recommendations on use (https://openscienceprobes.sgc-frankfurt.de/). Here we describe the chemical tools and target-related knowledge that have been made available, and encourage others to join the project.


Cancer Research | 2018

Abstract 1646: Discovery and characterization of BAY-6035, a novel benzodiazepine-based SMYD3 inhibitor

Stefan Gradl; Holger Steuber; Jörg Weiske; Norbert Schmees; Stephan Siegel; Detlef Stoeckigt; Clara D. Christ; Fengling Li; Shawna Organ; Dalia Barsyte-Lovejoy; Magdalena M. Szewczyk; Steven Kennedy; Viacheslav Trush; Masoud Vedadi; C.H. Arrowsmith; Peter J. Brown; Manfred Husemann; Amaury Ernesto Fernández-Montalván; Volker Badock; Marcus Bauser; Andrea Haegebarth; Ingo Hartung; Carlo Stresemann

SMYD3 (SET and MYND domain-containing protein 3) is a protein lysine methyltransferase (PKMT) which was initially described as H3K4 methyltransferase involved in transcriptional regulation. SMYD3 has recently been reported to methylate and regulate several non-histone cancer relevant proteins such as mitogen-activated protein kinase kinase kinase 2 (MAP3K2), vascular endothelial growth factor receptor 1 (VEGFR1), and the human epidermal growth factor receptor 2 (HER2). In addition overexpression of SMYD3 has been linked to poor prognosis in certain cancers, thus supporting a possible oncogenic role for SMYD3 and making it an attractive target for anticancer drug development. Here we report the discovery of a novel potent and selective SMYD3 inhibitor series. We performed a thermal shift assay based (TSA) high throughput screening followed by extensive biophysical validation resulting in identification of a benzodiazepine-based SMYD3 inhibitor series. The co-crystallization structures revealed that this series binds to the substrate binding site and occupies the hydrophobic pocket for lysine binding using an unprecedented hydrogen bond pattern. The competitive behavior of the inhibitor in biochemical assays was consistent with the binding mode observed in the crystal structure. Further optimization generated BAY-6035, which showed improved nanomolar potency and was selective against kinases and other PKMTs. Furthermore, BAY-6035 specifically inhibited methylation of MAP3K2 by SMYD3 in a cellular assay with similar potency. In summary, BAY-6035 is a novel selective and potent SMYD3 inhibitor probe and will foster the exploration of the biologic role of SMYD3 in diseased and non-diseased tissues. Citation Format: Stefan Gradl, Holger Steuber, Jorg Weiske, Norbert Schmees, Stephan Siegel, Detlef Stoeckigt, Clara D. Christ, Fengling Li, Shawna Organ, Dalia Barsyte-Lovejoy, Magdalena M. Szewczyk, Steven Kennedy, Viacheslav Trush, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Manfred Husemann, Amaury E. Fernandez-Montalvan, Volker Badock, Marcus Bauser, Andrea Haegebarth, Ingo V. Hartung, Carlo Stresemann. Discovery and characterization of BAY-6035, a novel benzodiazepine-based SMYD3 inhibitor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1646.


Cancer Research | 2016

Abstract 4746: Pharmacological characterization of BAY-876, a novel highly selective inhibitor of glucose transporter (GLUT)-1 in vitro and in vivo

Charlotte Kopitz; Luisella Toschi; Carolyn Algire; Mélanie Héroult; Anna-Lena Frisk; Kirstin Meyer; Arndt Schmitz; Eleni Lagkadinou; Heike Petrul; Iring Heisler; Roland Neuhaus; Bernd Buchmann; Herbert Himmel; Marcus Bauser; Andrea Haegebarth; Karl Ziegelbauer

One hallmark of cancer is the accelerated metabolism, high energy requirements, and increased glucose uptake by the tumor cells, the latter being the first and rate-limiting step for glucose metabolism. Glucose transport into the tumor cell is mediated by facilitative high-affinity glucose transporter (GLUT) proteins. Among the 14 GLUT proteins, expression of GLUT1 in normal organs is nearly exclusively restricted to the blood brain barrier, while other GLUTs are also expressed in a wide variety of vital organs such as liver and heart. Interestingly, GLUT1 expression is highly regulated by hypoxia-inducible factor (HIF)-1α, a key driver of tumor progression. In line with this finding, GLUT1 over-expression was found to be associated with tumor progression and poor overall survival in various tumor indications. Consequently, GLUT1 represents a potential target for cancer treatment. Therefore, we have developed a highly-selective GLUT1 inhibitor, namely BAY-876, with selectivity over GLUT2, 3, and 4 of 4700-, 800-, and 135-fold, respectively. We here show for the first time the pharmacological characterization of BAY-876, comprising inhibition of glucose-uptake, anti-proliferative activity in vitro, and anti-tumor efficacy in vivo in models of different tumor indications in monotherapy as well as first results on the combinability of BAY-876. Furthermore, at the therapeutic dose, BAY-876 treatment did not show any relevant finding on the behavior of treated mice in the Irwin test, assuming no or only minor effects on brain function. In conclusion, BAY-876 is the first GLUT1-selective inhibitor which reduces glucose uptake and growth of tumor cells with sufficient tolerability at the efficacious dose in preclinical models. Citation Format: Charlotte Kopitz, Luisella Toschi, Carolyn Algire, Melanie Heroult, Anna-Lena Frisk, Kirstin Meyer, Arndt Schmitz, Eleni Lagkadinou, Heike Petrul, Iring Heisler, Roland Neuhaus, Bernd Buchmann, Herbert Himmel, Marcus Bauser, Andrea Haegebarth, Karl Ziegelbauer. Pharmacological characterization of BAY-876, a novel highly selective inhibitor of glucose transporter (GLUT)-1 in vitro and in vivo. [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 4746.


Archive | 2000

Substituted 2-thio-3,5-dicyano-4-aryl-6-aminopyridines and the use thereof

Ulrich Rosentreter; Rolf Henning; Marcus Bauser; Thomas Dr. Krämer; Andrea Vaupel; Walter Dr. Hübsch; Klaus Dembowsky; Olga Salcher-Schraufstätter; Johannes-Peter Stasch; Thomas Krahn; Elisabeth Perzborn


Archive | 2008

Substituted oxazolidinones and their use

Swen Allerheiligen; Marcus Bauser; Hartmut Schirok; Ulrich Rester; Stefan Heitmeier; Christoph Gerdes; Georges Degenfeld; Elke Dittrich-Wengenroth; Uwe Saatmann; Julia Straβburger; Klaus Münter; Mark Jean Gnoth; Dieter Lang


Journal of Chromatographic Science | 2002

Preparative high-performance liquid chromatography-mass spectrometry for the high-throughput purification of combinatorial libraries

Marcus Bauser


Archive | 2008

Substituted (oxazolidinon-5-yl-methyl) -2-thiophene-carboxamides and use thereof in the field of blood coagulation

Swen Allerheiligen; Marcus Bauser; Dirk Heimbach; Stefan Heitmeier; Mark Jean Gnoth; Christoph Gerdes; Georges Degenfeld; Susanne Röhrig; Ulrich Rester; Elke Dittrich-Wengenroth; Uwe Saatmann; Adrian Tersteegen


Archive | 2001

Use of N-phenyl arylamide for treating or preventing chronic or acute hepatitis B viral infections in humans or animals, including co-infections with hepatitis D virus

Susanne Nikolic; Marcus Bauser; Michael Brands; Arnold Paeßens; Erwin Graef; Olaf Weber


ChemInform | 2005

Instrumentation for Combinatorial Chemistry

Marcus Bauser; Hubertus Stakemeier

Collaboration


Dive into the Marcus Bauser's collaboration.

Researchain Logo
Decentralizing Knowledge