Network


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

Hotspot


Dive into the research topics where Benjamin Bader is active.

Publication


Featured researches published by Benjamin Bader.


Molecular Cancer Therapeutics | 2016

Novel Mps1 Kinase Inhibitors with Potent Antitumor Activity.

Antje Margret Wengner; Gerhard Siemeister; Marcus Koppitz; Volker Schulze; Dirk Kosemund; Ulrich Klar; Detlef Stoeckigt; Roland Neuhaus; Philip Lienau; Benjamin Bader; Stefan Prechtl; Marian Raschke; Anna-Lena Frisk; Oliver von Ahsen; Martin Michels; Bertolt Kreft; Franz von Nussbaum; Michael Brands; Dominik Mumberg; Karl Ziegelbauer

Monopolar spindle 1 (Mps1) has been shown to function as the key kinase that activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we report the structure and functional characterization of two novel selective Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nmol/L while showing an excellent selectivity profile. In cellular mechanistic assays, both Mps1 inhibitors abrogated nocodazole-induced SAC activity and induced premature exit from mitosis (“mitotic breakthrough”), resulting in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 nmol/L range). In vivo, BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies. However, in line with its unique mode of action, when combined with paclitaxel, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest by the weakening of SAC activity. As a result, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor monotreatment at the respective MTDs in a broad range of xenograft models, including those showing acquired or intrinsic paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. These preclinical findings validate the innovative concept of SAC abrogation for cancer therapy and justify clinical proof-of-concept studies evaluating the Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs to enhance their efficacy and potentially overcome resistance. Mol Cancer Ther; 15(4); 583–92. ©2016 AACR.


Cancer Research | 2017

Abstract 983: Identification of potent, highly selective and orally available ATR inhibitor BAY 1895344 with favorable PK properties and promising efficacy in monotherapy and combination in preclinical tumor models

Ulrich Luecking; Julien Lefranc; Antje Margret Wengner; Lars Wortmann; Hans D. Schick; Hans Briem; Gerhard Siemeister; Philip Lienau; Christoph Schatz; Benjamin Bader; Gesa Deeg; Franz von Nussbaum; Michael Brands; Dominik Mumberg; Karl Ziegelbauer

The integrity of the genome of eukaryotic cells is secured by complex signaling pathways, known as DNA damage response (DDR). Recognition of DNA damage activates DDR pathways resulting in cell cycle arrest, suppression of general translation, induction of DNA repair, cell survival or even cell death. Proteins that directly recognize aberrant DNA structures recruit and activate kinases of the DDR pathway, such as ATR (ataxia telangiectasia and Rad3-related). ATR responds to a broad spectrum of DNA damage, including double-strand breaks (DSB) and lesions derived from interference with DNA replication as well as increased replication stress (e.g. in oncogene-driven tumor cells). Therefore, inhibition of ATR kinase activity could be the basis for a novel anti-cancer therapy in tumors with increased DNA damage, deficiency in DNA damage repair or replication stress. Herein we report the identification of the potent, highly selective and orally available ATR inhibitor BAY 1895344 by a collaborative effort involving medicinal chemistry, pharmacology, DMPK and computational chemistry. The chemical structures of lead compound BAY-937 and clinical candidate BAY 1895344 as well as the main SAR trends within this novel class of naphthyridine derivatives will be disclosed for the first time. The novel lead compound BAY-937 revealed promising inhibition of ATR (IC50 = 78 nM) and high kinase selectivity in vitro. In cellular mechanistic assays BAY-937 inhibited hydroxyurea-induced H2AX phosphorylation (IC50 = 380 nM) demonstrating the anticipated mode of action. Moreover, BAY-937 was shown to inhibit proliferation of a variety of tumor cell lines with low- to sub-micromolar IC50 values. In initial xenograft studies, BAY-937 revealed moderate activity in monotherapy and in combination with cis-platin. However, BAY-937 also revealed low aqueous solubility, low bioavailability (rat) and activity in the hERG patch clamp assay. Extensive lead optimization efforts led to the identification of the novel, orally available ATR inhibitor BAY 1895344. In vitro, BAY 1895344 was shown to be a very potent and highly selective ATR inhibitor (IC50 = 7 nM), which potently inhibits proliferation of a broad spectrum of human tumor cell lines (median IC50 = 78 nM). In cellular mechanistic assays BAY 1895344 potently inhibited hydroxyurea-induced H2AX phosphorylation (IC50 = 36 nM). Moreover, BAY 1895344 revealed significantly improved aqueous solubility, bioavailability across species and no activity in the hERG patch-clamp assay. BAY 1895344 also demonstrated very promising efficacy in monotherapy in DNA damage deficient tumor models as well as combination treatment with DNA damage inducing therapies. The start of clinical investigation of BAY 1895344 is planned for early 2017. Citation Format: Ulrich T. Luecking, Julien Lefranc, Antje Wengner, Lars Wortmann, Hans Schick, Hans Briem, Gerhard Siemeister, Philip Lienau, Christoph Schatz, Benjamin Bader, Gesa Deeg, Franz von Nussbaum, Michael Brands, Dominik Mumberg, Karl Ziegelbauer. Identification of potent, highly selective and orally available ATR inhibitor BAY 1895344 with favorable PK properties and promising efficacy in monotherapy and combination in preclinical tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 983. doi:10.1158/1538-7445.AM2017-983


Cancer Research | 2015

Abstract 3090: Novel Mps1 kinase inhibitors with potent anti-tumor activity

Antje Margret Wengner; Gerhard Siemeister; Marcus Koppitz; Volker Schulze; Dirk Kosemund; Ulrich Klar; Detlef Stoeckigt; Roland Neuhaus; Philip Lienau; Benjamin Bader; Stefan Prechtl; Olaf Doehr; Marian Raschke; Oliver von Ahsen; Cem Elbi; Ingmar Bruns; Martin Michels; Bertolt Kreft; Franz von Nussbaum; Michael Brands; Dominik Mumberg; Karl Ziegelbauer

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Cell cycle deregulation represents one of the hallmarks of cancer and consequently cell cycle arrest is the predominant mode of action for a number of antimitotic cancer drugs (e.g. taxanes and vinca alkaloids). Targeted disruption of the cell cycle checkpoint offers a novel approach to cancer treatment since tumor cells will not arrest in mitosis despite DNA damage or unattached/misattached chromosomes resulting in aneuploidy and cell death. Mps1, a mitotic kinase that is overexpressed in several human cancers, has been shown to function as the key kinase which activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we disclose for the first time the structure and functional characterization of two novel Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nM while showing an excellent selectivity profile against a broad panel of kinases. In cellular mechanistic assays, BAY 1161909 and BAY 1217389 abrogated nocodazole-induced SAC activity, inducing premature exit from mitosis (“mitotic breakthrough”), which results in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 values in low nanomolar range), showing a similar inhibitory pattern in a broad panel of tumor cell lines. In vivo, the Mps1 inhibitors BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies (tumor growth inhibition ∼ 50%). However, according to its unique mode of action, when combined with paclitaxel, at the maximum tolerated dose, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest in line with weakening of SAC activity. Consequently, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor mono-treatment in a broad range of xenograft models including those being intrinsically paclitaxel-insensitive as well as those with acquired paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. Our findings validate the innovative concept of SAC abrogation and justify clinical proof of concept studies evaluating Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs in order to enhance their efficacy and potentially overcome resistance. BAY 1161909 is currently in a phase I clinical trial ([NCT02138812][1]), start of clinical investigation of BAY 1217389 is planned. To our knowledge BAY 1161909 and BAY 1217389 are the first Mps1 inhibitors in clinical trials. Citation Format: Antje Margret Wengner, Gerhard Siemeister, Marcus Koppitz, Volker Schulze, Dirk Kosemund, Ulrich Klar, Detlef Stoeckigt, Roland Neuhaus, Philip Lienau, Benjamin Bader, Stefan Prechtl, Olaf Doehr, Marian Raschke, Oliver von Ahsen, Cem Elbi, Ingmar Bruns, Martin Michels, Bertolt Kreft, Franz von Nussbaum, Michael Brands, Dominik Mumberg, Karl Ziegelbauer. Novel Mps1 kinase inhibitors with potent anti-tumor activity. [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 3090. doi:10.1158/1538-7445.AM2015-3090 [1]: /lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT02138812&atom=%2Fcanres%2F75%2F15_Supplement%2F3090.atom


Cancer Research | 2014

Abstract 4540: SAC abrogation by MPS1 kinase inhibition: preclinical proof of concept of a novel approach to tumor treatment

Dominik Mumberg; Gerhard Siemeister; Antje Margret Wengner; Marcus Koppitz; Volker Schulze; Benjamin Bader; Stefan Prechtl; Bertolt Kreft; Karl Ziegelbauer

Cell cycle deregulation represents one of the classical hallmarks of cancer and consequently cell cycle arrest is the predominant mode of action of a number of antimitotic cancer drugs (e.g. taxanes and vinca alkaloids). Targeted disruption of the cell cycle checkpoint offers a novel approach to cancer treatment: driving tumor cells into cell division despite DNA damage or unattached/misattached chromosomes resulting in a lethal degree of DNA damage or aneuploidy. MPS1, a mitotic kinase that is overexpressed in several human cancers, has been shown to function as the key kinase which activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we report the identification and functional characterization of novel inhibitors of MPS1 from two structurally unrelated chemical classes. In biochemical assays, these molecules inhibited the kinase activity of MPS1 with IC50 values in the single digit nanomolar range and have shown an exquisite selectivity against a panel of kinases. In cellular assays the compounds were able to abrogate nocodazole-induced SAC, to reduce time in mitosis, to induce multinuclearity and apoptotic cell death, and to inhibit tumor cell proliferation with IC50 values in the low nanomolar range. In combination experiments MPS1 inhibitors showed cooperativity with low concentrations of paclitaxel. MPS1 inhibitors showed limited efficacy in monotherapy (T/C ca. 0.6) in tumor xenograft studies. However, when combined with paclitaxel dosed at the maximal tolerated dose, low doses of MPS1 inhibitor strongly improved efficacy over paclitaxel or MPS inhibitor monotreatment in intrinsically paclitaxel-insensitive xenograft models. The combination treatment was well tolerated. These results validate the concept of SAC abrogation preclinically and pave the way to a clinical proof of concept. Citation Format: Dominik Mumberg, Gerhard Siemeister, Antje M. Wengner, Marcus Koppitz, Volker Schulze, Benjamin Bader, Stefan Prechtl, Bertolt Kreft, Karl Ziegelbauer. SAC abrogation by MPS1 kinase inhibition: preclinical proof of concept of a novel approach to tumor treatment. [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 4540. doi:10.1158/1538-7445.AM2014-4540


Archive | 2006

Substituted imidazo[1,2b]pyridazines as kinase inhibitors, the production thereof, and use of the same as medicaments

Olaf Prien; Benjamin Bader; Ulrich Zügel; Stuart Ince; Christoph Huwe; Karina Schuck; Knut Eis; Ulrich Lücking; Rolf Jautelat; Judith Günther; Manfred Husemann


Archive | 2010

Substituted aminoquinoxalines as tyrosine threonine kinase inhibitors

Benjamin Bader; Ulf Bömer; Stuart Ince; Marcus Koppitz; Philip Lienau; Tobias Marquardt; Duy Nguyen; Stefan Prechtl; Gerhard Siemeister; Christof Wegscheid-Gerlach


Archive | 2007

OXO-SUBSTITUTED IMIDAZO[1,2b]PYRIDAZINES, PRODUCTION AND USE THEREOF AS DRUGS

Olaf Prien; Knut Eis; Benjamin Bader; Judith Guenther; Von Arne Bonin


Archive | 2013

BET-PROTEIN-INHIBITING DIHYDROPYRIDOPYRAZINONES

Norbert Schmees; Benjamin Bader; Bernard Haendler; Detlef Stöckigt; Pascale Lejeune; Amaury Ernesto Fernández-Montalván; Timo Stellfeld; Daniel Gallenkamp


Archive | 2006

(2,4,9-TRIAZA-1(2,4)-PYRIMIDINA-3(1,3)-BENZENACYCLONONAPHAN-3^4-YL)-SULFOXIMIDE DERIVATIVES AS SELECTIVE INHIBITORS OF THE AURORA KINASE FOR THE TREATMENT OF CANCER

Ulrich Lücking; Benjamin Bader; Gerhard Siemeister


Archive | 2016

SUBSTITUTED DIHYDROPYRIDO[3,4-B]PYRAZINONES AS DUAL INHIBITORS OF BET PROTEINS AND POLO-LIKE KINASES

Norbert Schmees; Benjamin Bader; Bernard Haendler; Volker Schulze; Ingo Hartung; Niels Böhnke; Florian Pühler

Collaboration


Dive into the Benjamin Bader's collaboration.

Top Co-Authors

Avatar

Gerhard Siemeister

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Philip Lienau

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Antje Margret Wengner

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Stefan Prechtl

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Volker Schulze

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Dominik Mumberg

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Karl Ziegelbauer

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Marcus Koppitz

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Bertolt Kreft

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Christoph Huwe

Bayer HealthCare Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge